51
|
Ockfen E, Filali L, Pereira Fernandes D, Hoffmann C, Thomas C. Actin cytoskeleton remodeling at the cancer cell side of the immunological synapse: good, bad, or both? Front Immunol 2023; 14:1276602. [PMID: 37869010 PMCID: PMC10585106 DOI: 10.3389/fimmu.2023.1276602] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 09/21/2023] [Indexed: 10/24/2023] Open
Abstract
Cytotoxic lymphocytes (CLs), specifically cytotoxic T lymphocytes and natural killer cells, are indispensable guardians of the immune system and orchestrate the recognition and elimination of cancer cells. Upon encountering a cancer cell, CLs establish a specialized cellular junction, known as the immunological synapse that stands as a pivotal determinant for effective cell killing. Extensive research has focused on the presynaptic side of the immunological synapse and elucidated the multiple functions of the CL actin cytoskeleton in synapse formation, organization, regulatory signaling, and lytic activity. In contrast, the postsynaptic (cancer cell) counterpart has remained relatively unexplored. Nevertheless, both indirect and direct evidence has begun to illuminate the significant and profound consequences of cytoskeletal changes within cancer cells on the outcome of the lytic immunological synapse. Here, we explore the understudied role of the cancer cell actin cytoskeleton in modulating the immune response within the immunological synapse. We shed light on the intricate interplay between actin dynamics and the evasion mechanisms employed by cancer cells, thus providing potential routes for future research and envisioning therapeutic interventions targeting the postsynaptic side of the immunological synapse in the realm of cancer immunotherapy. This review article highlights the importance of actin dynamics within the immunological synapse between cytotoxic lymphocytes and cancer cells focusing on the less-explored postsynaptic side of the synapse. It presents emerging evidence that actin dynamics in cancer cells can critically influence the outcome of cytotoxic lymphocyte interactions with cancer cells.
Collapse
Affiliation(s)
- Elena Ockfen
- Cytoskeleton and Cancer Progression, Department of Cancer Research, Luxembourg Institute of Health, Luxembourg, Luxembourg
- Faculty of Science, Technology and Medicine University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Liza Filali
- Cytoskeleton and Cancer Progression, Department of Cancer Research, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Diogo Pereira Fernandes
- Cytoskeleton and Cancer Progression, Department of Cancer Research, Luxembourg Institute of Health, Luxembourg, Luxembourg
- Faculty of Science, Technology and Medicine University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Céline Hoffmann
- Cytoskeleton and Cancer Progression, Department of Cancer Research, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Clément Thomas
- Cytoskeleton and Cancer Progression, Department of Cancer Research, Luxembourg Institute of Health, Luxembourg, Luxembourg
| |
Collapse
|
52
|
Ye Y, Morita S, Chang JJ, Buckley PM, Wilhelm KB, DiMaio D, Groves JT, Barrera FN. Allosteric inhibition of the T cell receptor by a designed membrane ligand. eLife 2023; 12:e82861. [PMID: 37796108 PMCID: PMC10554751 DOI: 10.7554/elife.82861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 09/20/2023] [Indexed: 10/06/2023] Open
Abstract
The T cell receptor (TCR) is a complex molecular machine that directs the activation of T cells, allowing the immune system to fight pathogens and cancer cells. Despite decades of investigation, the molecular mechanism of TCR activation is still controversial. One of the leading activation hypotheses is the allosteric model. This model posits that binding of pMHC at the extracellular domain triggers a dynamic change in the transmembrane (TM) domain of the TCR subunits, which leads to signaling at the cytoplasmic side. We sought to test this hypothesis by creating a TM ligand for TCR. Previously we described a method to create a soluble peptide capable of inserting into membranes and binding to the TM domain of the receptor tyrosine kinase EphA2 (Alves et al., eLife, 2018). Here, we show that the approach is generalizable to complex membrane receptors, by designing a TM ligand for TCR. We observed that the designed peptide caused a reduction of Lck phosphorylation of TCR at the CD3ζ subunit in T cells. As a result, in the presence of this peptide inhibitor of TCR (PITCR), the proximal signaling cascade downstream of TCR activation was significantly dampened. Co-localization and co-immunoprecipitation in diisobutylene maleic acid (DIBMA) native nanodiscs confirmed that PITCR was able to bind to the TCR. AlphaFold-Multimer predicted that PITCR binds to the TM region of TCR, where it interacts with the two CD3ζ subunits. Our results additionally indicate that PITCR disrupts the allosteric changes in the compactness of the TM bundle that occur upon TCR activation, lending support to the allosteric TCR activation model. The TCR inhibition achieved by PITCR might be useful to treat inflammatory and autoimmune diseases and to prevent organ transplant rejection, as in these conditions aberrant activation of TCR contributes to disease.
Collapse
Affiliation(s)
- Yujie Ye
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee at KnoxvilleKnoxvilleUnited States
| | - Shumpei Morita
- Department of Chemistry, University of California, BerkeleyBerkeleyUnited States
| | - Justin J Chang
- Department of Genetics, Yale UniversityNew HavenUnited States
| | - Patrick M Buckley
- Department of Microbial Pathogenesis, Yale UniversityNew HavenUnited States
| | - Kiera B Wilhelm
- Department of Chemistry, University of California, BerkeleyBerkeleyUnited States
| | - Daniel DiMaio
- Department of Genetics, Yale UniversityNew HavenUnited States
| | - Jay T Groves
- Department of Chemistry, University of California, BerkeleyBerkeleyUnited States
- Institute for Digital Molecular Analytics and Science, Nanyang Technological UniversitySingaporeSingapore
| | - Francisco N Barrera
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee at KnoxvilleKnoxvilleUnited States
| |
Collapse
|
53
|
Mandal S, Melo M, Gordiichuk P, Acharya S, Poh YC, Li N, Aung A, Dane EL, Irvine DJ, Kumari S. WASP facilitates tumor mechanosensitivity in T lymphocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.02.560434. [PMID: 37873483 PMCID: PMC10592916 DOI: 10.1101/2023.10.02.560434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Cytotoxic T lymphocytes (CTLs) carry out immunosurveillance by scanning target cells of diverse physical properties for the presence of antigens. While the recognition of cognate antigen by the T cell receptor is the primary signal for CTL activation, it has become increasingly clear that the mechanical stiffness of target cells plays an important role in antigen-triggered T cell responses. However, the molecular machinery within CTLs that transduces the mechanical information of tumor cells remains unclear. We find that CTL's mechanosensitive ability requires the activity of the actin-organizing protein Wiskott-Aldrich Syndrome Protein (WASP). WASP activation is modulated by the mechanical properties of antigen-presenting contexts across a wide range of target cell stiffnesses and activated WASP then mediates mechanosensitive activation of early TCR signaling markers in the CTL. Our results provide a molecular link between antigen mechanosensing and CTL immune response and suggest that CTL-intrinsic cytoskeletal organizing principles enable the processing of mechanical information from diverse target cells.
Collapse
Affiliation(s)
| | - Mariane Melo
- Koch Institute of Integrative Cancer Research, MIT, Cambridge, USA
| | | | | | - Yeh-Chuin Poh
- Koch Institute of Integrative Cancer Research, MIT, Cambridge, USA
| | - Na Li
- Koch Institute of Integrative Cancer Research, MIT, Cambridge, USA
| | - Aereas Aung
- Koch Institute of Integrative Cancer Research, MIT, Cambridge, USA
| | - Eric L. Dane
- Koch Institute of Integrative Cancer Research, MIT, Cambridge, USA
| | - Darrell J. Irvine
- Koch Institute of Integrative Cancer Research, MIT, Cambridge, USA
- Department of Biological Engineering, MIT, Cambridge, USA
- Howard Hughes Medical Institute, Ashburn, Virginia, USA
| | - Sudha Kumari
- Indian Institute of Science, Bengaluru, India
- Koch Institute of Integrative Cancer Research, MIT, Cambridge, USA
| |
Collapse
|
54
|
Abstract
T cell activation is initiated by the recognition of specific antigenic peptides and subsequently accomplished by complex signaling cascades. These aspects have been extensively studied for decades as pivotal factors in the establishment of adaptive immunity. However, how receptors or signaling molecules are organized in the resting state prior to encountering antigens has received less attention. Recent advancements in super-resolution microscopy techniques have revealed topographically controlled pre-formed organization of key molecules involved in antigen recognition and signal transduction on microvillar projections of T cells before activation and substantial effort has been dedicated to characterizing the topological structure of resting T cells over the past decade. This review will summarize our current understanding of how key surface receptors are pre-organized on the T-cell plasma membrane and discuss the potential role of these receptors, which are preassembled prior to ligand binding in the early activation events of T cells.
Collapse
Affiliation(s)
- Yunmin Jung
- Department of Nano-Biomedical Engineering, Advanced Science Institute, Yonsei University, Seoul, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science, Seoul, Republic of Korea
| |
Collapse
|
55
|
Sajman J, Yakovian O, Unger Deshet N, Almog S, Horn G, Waks T, Globerson Levin A, Sherman E. Nanoscale CAR Organization at the Immune Synapse Correlates with CAR-T Effector Functions. Cells 2023; 12:2261. [PMID: 37759484 PMCID: PMC10527520 DOI: 10.3390/cells12182261] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/03/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
T cells expressing chimeric antigen receptors (CARs) are at the forefront of clinical treatment of cancers. Still, the nanoscale organization of CARs at the interface of CAR-Ts with target cells, which is essential for TCR-mediated T cell activation, remains poorly understood. Here, we studied the nanoscale organization of CARs targeting CD138 proteoglycans in such fixed and live interfaces, generated optimally for single-molecule localization microscopy. CARs showed significant self-association in nanoclusters that was enhanced in interfaces with on-target cells (SKOV-3, CAG, FaDu) relative to negative cells (OVCAR-3). CARs also segregated more efficiently from the abundant membrane phosphatase CD45 in CAR-T cells forming such interfaces. CAR clustering and segregation from CD45 correlated with the effector functions of Ca++ influx and target cell killing. Our results shed new light on the nanoscale organization of CARs on the surfaces of CAR-Ts engaging on- and off-target cells, and its potential significance for CAR-Ts' efficacy and safety.
Collapse
Affiliation(s)
- Julia Sajman
- Racah Institute of Physics, The Hebrew University, Jerusalem 91904, Israel
- Jerusalem College of Technology, Jerusalem 91160, Israel
| | - Oren Yakovian
- Racah Institute of Physics, The Hebrew University, Jerusalem 91904, Israel
| | - Naamit Unger Deshet
- Immunology and Advanced CAR-T Cell Therapy Laboratory, Research & Development Department, Tel-Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel
| | - Shaked Almog
- Immunology and Advanced CAR-T Cell Therapy Laboratory, Research & Development Department, Tel-Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel
| | - Galit Horn
- Immunology and Advanced CAR-T Cell Therapy Laboratory, Research & Development Department, Tel-Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel
| | - Tova Waks
- Immunology and Advanced CAR-T Cell Therapy Laboratory, Research & Development Department, Tel-Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel
| | - Anat Globerson Levin
- Immunology and Advanced CAR-T Cell Therapy Laboratory, Research & Development Department, Tel-Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel
- Dotan Center for Advanced Therapies, Tel-Aviv Sourasky Medical Center and Tel Aviv University, Tel Aviv 6423906, Israel
| | - Eilon Sherman
- Racah Institute of Physics, The Hebrew University, Jerusalem 91904, Israel
| |
Collapse
|
56
|
Wang T, Shi S, Shi Y, Jiang P, Hu G, Ye Q, Shi Z, Yu K, Wang C, Fan G, Zhao S, Ma H, Chang ACY, Li Z, Bian Q, Lin CP. Chemical-induced phase transition and global conformational reorganization of chromatin. Nat Commun 2023; 14:5556. [PMID: 37689690 PMCID: PMC10492836 DOI: 10.1038/s41467-023-41340-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 08/29/2023] [Indexed: 09/11/2023] Open
Abstract
Chemicals or drugs can accumulate within biomolecular condensates formed through phase separation in cells. Here, we use super-resolution imaging to search for chemicals that induce phase transition within chromatin at the microscale. This microscopic screening approach reveals that adriamycin (doxorubicin) - a widely used anticancer drug that is known to interact with chromatin - specifically induces visible local condensation and global conformational change of chromatin in cancer and primary cells. Hi-C and ATAC-seq experiments systematically and quantitatively demonstrate that adriamycin-induced chromatin condensation is accompanied by weakened chromatin interaction within topologically associated domains, compartment A/B switching, lower chromatin accessibility, and corresponding transcriptomic changes. Mechanistically, adriamycin complexes with histone H1 and induces phase transition of H1, forming fibrous aggregates in vitro. These results reveal a phase separation-driven mechanism for a chemotherapeutic drug.
Collapse
Affiliation(s)
- Tengfei Wang
- School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China
| | - Shuxiang Shi
- School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China
- Lingang Laboratory, 200031, Shanghai, China
| | - Yuanyuan Shi
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 200125, Shanghai, China
- Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Peipei Jiang
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 200125, Shanghai, China
- Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Ganlu Hu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, 201210, Shanghai, China
| | - Qinying Ye
- School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China
| | - Zhan Shi
- School of Physical Science and Technology, ShanghaiTech University, 201210, Shanghai, China
| | - Kexin Yu
- School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China
- iHuman Institute, ShanghaiTech University, 201010, Shanghai, China
| | - Chenguang Wang
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 200125, Shanghai, China
- Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
- Department of Cardiology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guoping Fan
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, 201210, Shanghai, China
| | - Suwen Zhao
- School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China
- iHuman Institute, ShanghaiTech University, 201010, Shanghai, China
| | - Hanhui Ma
- School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China
| | - Alex C Y Chang
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 200125, Shanghai, China
- Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
- Department of Cardiology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhi Li
- School of Physical Science and Technology, ShanghaiTech University, 201210, Shanghai, China
| | - Qian Bian
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 200125, Shanghai, China.
- Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.
| | - Chao-Po Lin
- School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China.
| |
Collapse
|
57
|
Sapoznikov A, Kozlovski S, Levi N, Feigelson SW, Regev O, Davidzohn N, Ben-Dor S, Haffner-Krausz R, Feldmesser E, Wigoda N, Petrovich-Kopitman E, Biton M, Alon R. Dendritic cell ICAM-1 strengthens synapses with CD8 T cells but is not required for their early differentiation. Cell Rep 2023; 42:112864. [PMID: 37494182 DOI: 10.1016/j.celrep.2023.112864] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 06/13/2023] [Accepted: 07/10/2023] [Indexed: 07/28/2023] Open
Abstract
Lymphocyte priming in lymph nodes (LNs) was postulated to depend on the formation of stable T cell receptor (TCR)-specific immune synapses (ISs) with antigen (Ag)-presenting dendritic cells (DCs). The high-affinity LFA-1 ligand ICAM-1 was implicated in different ISs studied in vitro. We dissect the in vivo roles of endogenous DC ICAM-1 in Ag-stimulated T cell proliferation and differentiation and find that under type 1 polarizing conditions in vaccinated or vaccinia virus-infected skin-draining LNs, Ag-presenting DCs engage in ICAM-1-dependent stable conjugates with a subset of Ag-specific CD8 blasts. Nevertheless, in the absence of these conjugates, CD8 lymphocyte proliferation and differentiation into functional cytotoxic T cells (CTLs) and skin homing effector lymphocytes takes place normally. Our results suggest that although CD8 T cell blasts engage in tight ICAM-1-dependent DC-T ISs, firm ISs are dispensable for TCR-triggered proliferation and differentiation into productive effector lymphocytes.
Collapse
Affiliation(s)
- Anita Sapoznikov
- Deptartment of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Stav Kozlovski
- Deptartment of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Nehora Levi
- Deptartment of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Sara W Feigelson
- Deptartment of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ofer Regev
- Deptartment of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Natalia Davidzohn
- Deptartment of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Shifra Ben-Dor
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | | | - Ester Feldmesser
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Noa Wigoda
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | | | - Moshe Biton
- Deptartment of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel.
| | - Ronen Alon
- Deptartment of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
58
|
Worboys JD, Vowell KN, Hare RK, Ambrose AR, Bertuzzi M, Conner MA, Patel FP, Zammit WH, Gali-Moya J, Hazime KS, Jones KL, Rey C, Jonjic S, Rovis TL, Tannahill GM, Cruz De Matos GDS, Waight JD, Davis DM. TIGIT can inhibit T cell activation via ligation-induced nanoclusters, independent of CD226 co-stimulation. Nat Commun 2023; 14:5016. [PMID: 37596248 PMCID: PMC10439114 DOI: 10.1038/s41467-023-40755-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 08/09/2023] [Indexed: 08/20/2023] Open
Abstract
TIGIT is an inhibitory receptor expressed on lymphocytes and can inhibit T cells by preventing CD226 co-stimulation through interactions in cis or through competition of shared ligands. Whether TIGIT directly delivers cell-intrinsic inhibitory signals in T cells remains unclear. Here we show, by analysing lymphocytes from matched human tumour and peripheral blood samples, that TIGIT and CD226 co-expression is rare on tumour-infiltrating lymphocytes. Using super-resolution microscopy and other techniques, we demonstrate that ligation with CD155 causes TIGIT to reorganise into dense nanoclusters, which coalesce with T cell receptor (TCR)-rich clusters at immune synapses. Functionally, this reduces cytokine secretion in a manner dependent on TIGIT's intracellular ITT-like signalling motif. Thus, we provide evidence that TIGIT directly inhibits lymphocyte activation, acting independently of CD226, requiring intracellular signalling that is proximal to the TCR. Within the subset of tumours where TIGIT-expressing cells do not commonly co-express CD226, this will likely be the dominant mechanism of action.
Collapse
Affiliation(s)
- Jonathan D Worboys
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | | | - Roseanna K Hare
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Ashley R Ambrose
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Margherita Bertuzzi
- Manchester Fungal Infection Group, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | | | | | - William H Zammit
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Judit Gali-Moya
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
- Department of Life Sciences, Sir Alexander Fleming Building, Imperial College London, South Kensington, London, UK
| | - Khodor S Hazime
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
- Department of Life Sciences, Sir Alexander Fleming Building, Imperial College London, South Kensington, London, UK
| | - Katherine L Jones
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Camille Rey
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Stipan Jonjic
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Tihana Lenac Rovis
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | | | | | | | - Daniel M Davis
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK.
- Department of Life Sciences, Sir Alexander Fleming Building, Imperial College London, South Kensington, London, UK.
| |
Collapse
|
59
|
Lan C, Kim J, Ulferts S, Aprile-Garcia F, Weyrauch S, Anandamurugan A, Grosse R, Sawarkar R, Reinhardt A, Hugel T. Quantitative real-time in-cell imaging reveals heterogeneous clusters of proteins prior to condensation. Nat Commun 2023; 14:4831. [PMID: 37582808 PMCID: PMC10427612 DOI: 10.1038/s41467-023-40540-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 07/26/2023] [Indexed: 08/17/2023] Open
Abstract
Our current understanding of biomolecular condensate formation is largely based on observing the final near-equilibrium condensate state. Despite expectations from classical nucleation theory, pre-critical protein clusters were recently shown to form under subsaturation conditions in vitro; if similar long-lived clusters comprising more than a few molecules are also present in cells, our understanding of the physical basis of biological phase separation may fundamentally change. Here, we combine fluorescence microscopy with photobleaching analysis to quantify the formation of clusters of NELF proteins in living, stressed cells. We categorise small and large clusters based on their dynamics and their response to p38 kinase inhibition. We find a broad distribution of pre-condensate cluster sizes and show that NELF protein cluster formation can be explained as non-classical nucleation with a surprisingly flat free-energy landscape for a wide range of sizes and an inhibition of condensation in unstressed cells.
Collapse
Affiliation(s)
- Chenyang Lan
- Institute of Physical Chemistry, University of Freiburg, Freiburg, Germany
- BIOSS and CIBSS Signalling Research Centres, University of Freiburg, Freiburg, Germany
- PicoQuant GmbH, Rudower Chaussee 29, 12489, Berlin, Germany
| | - Juhyeong Kim
- Institute of Physical Chemistry, University of Freiburg, Freiburg, Germany
| | - Svenja Ulferts
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Freiburg, Freiburg, Germany
| | | | - Sophie Weyrauch
- Institute of Physical Chemistry, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany
- Faculty of Chemistry and Pharmacology, University of Freiburg, Freiburg, Germany
| | | | - Robert Grosse
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Freiburg, Freiburg, Germany
| | - Ritwick Sawarkar
- Medical Research Council (MRC), University of Cambridge, Cambridge, CB2 1QR, United Kingdom
| | - Aleks Reinhardt
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, United Kingdom.
| | - Thorsten Hugel
- Institute of Physical Chemistry, University of Freiburg, Freiburg, Germany.
- BIOSS and CIBSS Signalling Research Centres, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
60
|
Bertolini M, Wong MS, Mendive-Tapia L, Vendrell M. Smart probes for optical imaging of T cells and screening of anti-cancer immunotherapies. Chem Soc Rev 2023; 52:5352-5372. [PMID: 37376918 PMCID: PMC10424634 DOI: 10.1039/d2cs00928e] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Indexed: 06/29/2023]
Abstract
T cells are an essential part of the immune system with crucial roles in adaptive response and the maintenance of tissue homeostasis. Depending on their microenvironment, T cells can be differentiated into multiple states with distinct functions. This myriad of cellular activities have prompted the development of numerous smart probes, ranging from small molecule fluorophores to nanoconstructs with variable molecular architectures and fluorescence emission mechanisms. In this Tutorial Review, we summarize recent efforts in the design, synthesis and application of smart probes for imaging T cells in tumors and inflammation sites by targeting metabolic and enzymatic biomarkers as well as specific surface receptors. Finally, we briefly review current strategies for how smart probes are employed to monitor the response of T cells to anti-cancer immunotherapies. We hope that this Review may help chemists, biologists and immunologists to design the next generation of molecular imaging probes for T cells and anti-cancer immunotherapies.
Collapse
Affiliation(s)
- Marco Bertolini
- Centre for Inflammation Research, The University of Edinburgh, EH16 4UU, Edinburgh, UK.
- IRR Chemistry Hub, Institute for Regeneration and Repair, The University of Edinburgh, EH16 4UU, Edinburgh, UK
| | - Man Sing Wong
- Centre for Inflammation Research, The University of Edinburgh, EH16 4UU, Edinburgh, UK.
- IRR Chemistry Hub, Institute for Regeneration and Repair, The University of Edinburgh, EH16 4UU, Edinburgh, UK
| | - Lorena Mendive-Tapia
- Centre for Inflammation Research, The University of Edinburgh, EH16 4UU, Edinburgh, UK.
- IRR Chemistry Hub, Institute for Regeneration and Repair, The University of Edinburgh, EH16 4UU, Edinburgh, UK
| | - Marc Vendrell
- Centre for Inflammation Research, The University of Edinburgh, EH16 4UU, Edinburgh, UK.
- IRR Chemistry Hub, Institute for Regeneration and Repair, The University of Edinburgh, EH16 4UU, Edinburgh, UK
| |
Collapse
|
61
|
Miao Y, Guo X, Zhu K, Zhao W. Biomolecular condensates tunes immune signaling at the Host-Pathogen interface. CURRENT OPINION IN PLANT BIOLOGY 2023; 74:102374. [PMID: 37148673 DOI: 10.1016/j.pbi.2023.102374] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 04/06/2023] [Accepted: 04/07/2023] [Indexed: 05/08/2023]
Abstract
Membraneless organelles participate in diverse spatiotemporal regulation of cellular signal transduction by recruiting necessary signaling factors. During host-pathogen interactions, the plasma membrane (PM) at the interface between the plant and microbes serves as a central platform for forming multicomponent immune signaling hubs. The macromolecular condensation of the immune complex and regulators is important in regulating immune signaling outputs regarding strength, timing, and crosstalk between signaling pathways. This review discusses mechanisms that regulate specific and crosstalk of plant immune signal transduction pathways through macromolecular assembly and condensation.
Collapse
Affiliation(s)
- Yansong Miao
- School of Biological Sciences, Nanyang Technological University, 637551, Singapore; Institute for Digital Molecular Analytics and Science, Nanyang Technological University, 636921, Singapore.
| | - Xiangfu Guo
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, 637457, Singapore
| | - Kexin Zhu
- School of Biological Sciences, Nanyang Technological University, 637551, Singapore
| | - Wenting Zhao
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, 637457, Singapore; Institute for Digital Molecular Analytics and Science, Nanyang Technological University, 636921, Singapore
| |
Collapse
|
62
|
Barr VA, Piao J, Balagopalan L, McIntire KM, Schoenberg FP, Samelson LE. Heterogeneity of Signaling Complex Nanostructure in T Cells Activated Via the T Cell Antigen Receptor. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2023; 29:1503-1522. [PMID: 37488826 PMCID: PMC11230849 DOI: 10.1093/micmic/ozad072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 06/08/2023] [Accepted: 06/18/2023] [Indexed: 07/26/2023]
Abstract
Activation of the T cell antigen receptor (TCR) is a key step in initiating the adaptive immune response. Single-molecule localization techniques have been used to investigate the arrangement of proteins within the signaling complexes formed around activated TCRs, but a clear picture of nanoscale organization in stimulated T cells has not emerged. Here, we have improved the examination of T cell nanostructure by visualizing individual molecules of six different proteins in a single sample of activated Jurkat T cells using the multiplexed antibody-size limited direct stochastic optical reconstruction microscopy (madSTORM) technique. We formally define irregularly shaped regions of interest, compare areas where signaling complexes are concentrated with other areas, and improve the statistical analyses of the locations of molecules. We show that nanoscale organization of proteins is mainly confined to the areas with dense concentrations of TCR-based signaling complexes. However, randomly distributed molecules are also found in some areas containing concentrated signaling complexes. These results are consistent with the view that the proteins within signaling complexes are connected by numerous weak interactions, leading to flexible, dynamic, and mutable structures which produce large variations in the nanostructure found in activated T cells.
Collapse
Affiliation(s)
- Valarie A Barr
- Laboratory of Cellular & Molecular Biology, Building 37 Room 2066, 37 Convent Drive, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892-4256, USA
| | - Juan Piao
- Department of Statistics, University of California at Los Angeles, 8965 Math Sciences Building, Los Angeles, CA 90095-1554, USA
| | - Lakshmi Balagopalan
- Laboratory of Cellular & Molecular Biology, Building 37 Room 2066, 37 Convent Drive, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892-4256, USA
| | - Katherine M McIntire
- Laboratory of Cellular & Molecular Biology, Building 37 Room 2066, 37 Convent Drive, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892-4256, USA
| | - Frederic P Schoenberg
- Department of Statistics, University of California at Los Angeles, 8965 Math Sciences Building, Los Angeles, CA 90095-1554, USA
| | - Lawrence E Samelson
- Laboratory of Cellular & Molecular Biology, Building 37 Room 2066, 37 Convent Drive, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892-4256, USA
| |
Collapse
|
63
|
Liang Q, Wang L, Xu J, Lin A, Wu Y, Tao Q, Zhang B, Min H, Song S, Gao Q. A burns and COVID-19 shared stress responding gene network deciphers CD1C-CD141- DCs as the key cellular components in septic prognosis. Cell Death Discov 2023; 9:258. [PMID: 37488118 PMCID: PMC10366195 DOI: 10.1038/s41420-023-01518-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/15/2023] [Accepted: 06/22/2023] [Indexed: 07/26/2023] Open
Abstract
Differential body responses to various stresses, infectious or noninfectious, govern clinical outcomes ranging from asymptoma to death. However, the common molecular and cellular nature of the stress responsome across different stimuli is not described. In this study, we compared the expression behaviors between burns and COVID-19 infection by choosing the transcriptome of peripheral blood from related patients as the analytic target since the blood cells reflect the systemic landscape of immune status. To this end, we identified an immune co-stimulator (CD86)-centered network, named stress-response core (SRC), which was robustly co-expressed in burns and COVID-19. The enhancement of SRC genes (SRCs) expression indicated favorable prognosis and less severity in both conditions. An independent whole blood single-cell RNA sequencing of COVID-19 patients demonstrated that the monocyte-dendritic cell (Mono-DC) wing was the major cellular source of SRC, among which the higher expression of the SRCs in the monocyte was associated with the asymptomatic COVID-19 patients, while the quantity-restricted and function-defected CD1C-CD141-DCs were recognized as the key signature which linked to bad consequences. Specifically, the proportion of the CD1C-CD141-DCs and their SRCs expression were step-wise reduced along with worse clinic conditions while the subcluster of CD1C-CD141-DCs from the critical COVID-19 patients was characterized of IFN signaling quiescence, high mitochondrial metabolism and immune-communication inactivation. Thus, our study identified an expression-synchronized and function-focused gene network in Mono-DC population whose expression status was prognosis-related and might serve as a new target of diagnosis and therapy.
Collapse
Affiliation(s)
- Qiao Liang
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, Jiangsu Province, China
| | - Lei Wang
- Department of Clinical Laboratory, Jiangsu Provincial Hospital of Integrated Chinese and Western Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, China
| | - Jing Xu
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, Jiangsu Province, China
| | - Anqi Lin
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, Jiangsu Province, China
| | - Yongzheng Wu
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, Jiangsu Province, China
| | - Qing Tao
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, Jiangsu Province, China
| | - Bin Zhang
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, Jiangsu Province, China
- Central Laboratory, Nanjing Chest Hospital, Nanjing Medical University, Nanjing, 210028, China
| | - Haiyan Min
- Central Laboratory, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210028, China
| | - Shiyu Song
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, Jiangsu Province, China.
| | - Qian Gao
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, Jiangsu Province, China.
| |
Collapse
|
64
|
Guo X, Zhu K, Zhu X, Zhao W, Miao Y. Two-dimensional molecular condensation in cell signaling and mechanosensing. Acta Biochim Biophys Sin (Shanghai) 2023; 55:1064-1074. [PMID: 37475548 PMCID: PMC10423693 DOI: 10.3724/abbs.2023132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 05/21/2023] [Indexed: 07/22/2023] Open
Abstract
Membraneless organelles (MLO) regulate diverse biological processes in a spatiotemporally controlled manner spanning from inside to outside of the cells. The plasma membrane (PM) at the cell surface serves as a central platform for forming multi-component signaling hubs that sense mechanical and chemical cues during physiological and pathological conditions. During signal transduction, the assembly and formation of membrane-bound MLO are dynamically tunable depending on the physicochemical properties of the surrounding environment and partitioning biomolecules. Biomechanical properties of MLO-associated membrane structures can control the microenvironment for biomolecular interactions and assembly. Lipid-protein complex interactions determine the catalytic region's assembly pattern and assembly rate and, thereby, the amplitude of activities. In this review, we will focus on how cell surface microenvironments, including membrane curvature, surface topology and tension, lipid-phase separation, and adhesion force, guide the assembly of PM-associated MLO for cell signal transductions.
Collapse
Affiliation(s)
- Xiangfu Guo
- School of ChemistryChemical Engineering and BiotechnologyNanyang Technological UniversitySingapore637457Singapore
| | - Kexin Zhu
- School of Biological SciencesNanyang Technological UniversitySingapore637551Singapore
| | - Xinlu Zhu
- School of Biological SciencesNanyang Technological UniversitySingapore637551Singapore
| | - Wenting Zhao
- School of ChemistryChemical Engineering and BiotechnologyNanyang Technological UniversitySingapore637457Singapore
- Institute for Digital Molecular Analytics and ScienceNanyang Technological UniversitySingapore636921Singapore
| | - Yansong Miao
- School of Biological SciencesNanyang Technological UniversitySingapore637551Singapore
- Institute for Digital Molecular Analytics and ScienceNanyang Technological UniversitySingapore636921Singapore
| |
Collapse
|
65
|
Haake M, Haack B, Schäfer T, Harter PN, Mattavelli G, Eiring P, Vashist N, Wedekink F, Genssler S, Fischer B, Dahlhoff J, Mokhtari F, Kuzkina A, Welters MJP, Benz TM, Sorger L, Thiemann V, Almanzar G, Selle M, Thein K, Späth J, Gonzalez MC, Reitinger C, Ipsen-Escobedo A, Wistuba-Hamprecht K, Eichler K, Filipski K, Zeiner PS, Beschorner R, Goedemans R, Gogolla FH, Hackl H, Rooswinkel RW, Thiem A, Roche PR, Joshi H, Pühringer D, Wöckel A, Diessner JE, Rüdiger M, Leo E, Cheng PF, Levesque MP, Goebeler M, Sauer M, Nimmerjahn F, Schuberth-Wagner C, von Felten S, Mittelbronn M, Mehling M, Beilhack A, van der Burg SH, Riedel A, Weide B, Dummer R, Wischhusen J. Tumor-derived GDF-15 blocks LFA-1 dependent T cell recruitment and suppresses responses to anti-PD-1 treatment. Nat Commun 2023; 14:4253. [PMID: 37474523 PMCID: PMC10359308 DOI: 10.1038/s41467-023-39817-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 06/09/2023] [Indexed: 07/22/2023] Open
Abstract
Immune checkpoint blockade therapy is beneficial and even curative for some cancer patients. However, the majority don't respond to immune therapy. Across different tumor types, pre-existing T cell infiltrates predict response to checkpoint-based immunotherapy. Based on in vitro pharmacological studies, mouse models and analyses of human melanoma patients, we show that the cytokine GDF-15 impairs LFA-1/β2-integrin-mediated adhesion of T cells to activated endothelial cells, which is a pre-requisite of T cell extravasation. In melanoma patients, GDF-15 serum levels strongly correlate with failure of PD-1-based immune checkpoint blockade therapy. Neutralization of GDF-15 improves both T cell trafficking and therapy efficiency in murine tumor models. Thus GDF-15, beside its known role in cancer-related anorexia and cachexia, emerges as a regulator of T cell extravasation into the tumor microenvironment, which provides an even stronger rationale for therapeutic anti-GDF-15 antibody development.
Collapse
Affiliation(s)
- Markus Haake
- Department of Gynecology, University Hospital Würzburg, Würzburg, Germany
- CatalYm GmbH, Am Klopferspitz 19, 82152, Munich, Germany
| | - Beatrice Haack
- Department of Gynecology, University Hospital Würzburg, Würzburg, Germany
| | - Tina Schäfer
- Department of Gynecology, University Hospital Würzburg, Würzburg, Germany
| | - Patrick N Harter
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Neurological Institute (Edinger Institute), University Hospital, Goethe University, Frankfurt/Main, Germany
- Frankfurt Cancer Institute (FCI), Frankfurt/Main, Germany
- Center for Neuropathology and Prion Research, Munich, Ludwig-Maximilians-University, Munich, Germany
| | - Greta Mattavelli
- Mildred Scheel Early Career Center, University Hospital of Würzburg, Würzburg, Germany
| | - Patrick Eiring
- Department of Biotechnology and Biophysics, Julius Maximilians University Würzburg, Am Hubland, 97074, Würzburg, Germany
| | - Neha Vashist
- Department of Gynecology, University Hospital Würzburg, Würzburg, Germany
- CatalYm GmbH, Am Klopferspitz 19, 82152, Munich, Germany
| | - Florian Wedekink
- Department of Gynecology, University Hospital Würzburg, Würzburg, Germany
| | | | - Birgitt Fischer
- Department of Gynecology, University Hospital Würzburg, Würzburg, Germany
- CatalYm GmbH, Am Klopferspitz 19, 82152, Munich, Germany
| | - Julia Dahlhoff
- Department of Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | - Fatemeh Mokhtari
- Department of Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | - Anastasia Kuzkina
- Department of Gynecology, University Hospital Würzburg, Würzburg, Germany
| | - Marij J P Welters
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Albinusdreef 2, Leiden, 2333 ZA, The Netherlands
| | - Tamara M Benz
- Department of Gynecology, University Hospital Würzburg, Würzburg, Germany
| | - Lena Sorger
- Department of Gynecology, University Hospital Würzburg, Würzburg, Germany
| | - Vincent Thiemann
- Department of Gynecology, University Hospital Würzburg, Würzburg, Germany
| | - Giovanni Almanzar
- Department of Gynecology, University Hospital Würzburg, Würzburg, Germany
- Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
| | - Martina Selle
- Department of Gynecology, University Hospital Würzburg, Würzburg, Germany
| | - Klara Thein
- Department of Gynecology, University Hospital Würzburg, Würzburg, Germany
| | - Jacob Späth
- Department of Gynecology, University Hospital Würzburg, Würzburg, Germany
| | | | - Carmen Reitinger
- Division of Genetics, Department of Biology, University of Erlangen, 91058, Erlangen, Germany
| | - Andrea Ipsen-Escobedo
- Division of Genetics, Department of Biology, University of Erlangen, 91058, Erlangen, Germany
| | - Kilian Wistuba-Hamprecht
- Department of Dermatology, University Medical Center Tübingen, Tübingen, Germany
- Department of Immunology, University of Tübingen, Tübingen, Germany
- Section for Clinical Bioinformatics, Department of Internal Medicine I, University Medical Center Tübingen, Tübingen, Germany
| | - Kristin Eichler
- Department of Gynecology, University Hospital Würzburg, Würzburg, Germany
- CatalYm GmbH, Am Klopferspitz 19, 82152, Munich, Germany
| | - Katharina Filipski
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Neurological Institute (Edinger Institute), University Hospital, Goethe University, Frankfurt/Main, Germany
- Frankfurt Cancer Institute (FCI), Frankfurt/Main, Germany
| | - Pia S Zeiner
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Neurological Institute (Edinger Institute), University Hospital, Goethe University, Frankfurt/Main, Germany
- Frankfurt Cancer Institute (FCI), Frankfurt/Main, Germany
- Dr. Senckenberg Institute of Neurooncology, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Rudi Beschorner
- Department of Neuropathology, University of Tübingen, Tübingen, Germany
| | - Renske Goedemans
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Albinusdreef 2, Leiden, 2333 ZA, The Netherlands
| | - Falk Hagen Gogolla
- Institute of Bioinformatics, Biocenter, Medical University of Innsbruck, Innrain 80, 6020, Innsbruck, Austria
| | - Hubert Hackl
- Institute of Bioinformatics, Biocenter, Medical University of Innsbruck, Innrain 80, 6020, Innsbruck, Austria
| | | | - Alexander Thiem
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Würzburg, Germany
- Clinic for Dermatology and Venereology, Rostock University Medical Center, Rostock, Germany
| | - Paula Romer Roche
- Department of Gynecology, University Hospital Würzburg, Würzburg, Germany
- CatalYm GmbH, Am Klopferspitz 19, 82152, Munich, Germany
| | - Hemant Joshi
- Department of Gynecology, University Hospital Würzburg, Würzburg, Germany
- Division of Infectious Diseases, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, 63130, USA
| | - Dirk Pühringer
- Department of Gynecology, University Hospital Würzburg, Würzburg, Germany
| | - Achim Wöckel
- Department of Gynecology, University Hospital Würzburg, Würzburg, Germany
| | - Joachim E Diessner
- Department of Gynecology, University Hospital Würzburg, Würzburg, Germany
| | | | - Eugen Leo
- CatalYm GmbH, Am Klopferspitz 19, 82152, Munich, Germany
| | - Phil F Cheng
- Department of Dermatology, University of Zurich, University of Zurich Hospital, Wagistrasse 18, 8952, Zürich, Switzerland
| | - Mitchell P Levesque
- Department of Dermatology, University of Zurich, University of Zurich Hospital, Wagistrasse 18, 8952, Zürich, Switzerland
| | - Matthias Goebeler
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Würzburg, Germany
| | - Markus Sauer
- Department of Biotechnology and Biophysics, Julius Maximilians University Würzburg, Am Hubland, 97074, Würzburg, Germany
| | - Falk Nimmerjahn
- Division of Genetics, Department of Biology, University of Erlangen, 91058, Erlangen, Germany
| | | | - Stefanie von Felten
- oikostat GmbH, Statistical Analyses and Consulting, Lucerne, Switzerland
- Department of Biostatistics, Epidemiology, Biostatistics and Prevention Institute, University of Zurich, Hirschengraben 84, 8001, Zürich, Switzerland
| | - Michel Mittelbronn
- Department of Oncology (DONC), Luxembourg Institute of Health (LIH), Luxembourg, Luxembourg
- Luxembourg Centre of Neuropathology (LCNP), Luxembourg, Luxembourg
- National Center of Pathology (NCP), Laboratoire National de Santé (LNS), Dudelange, Luxembourg
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, Luxembourg, Luxembourg
- Faculty of Science, Technology and Medicine (FSTM), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Matthias Mehling
- Department of Biomedicine and Neurology Department, University Hospital Basel, 4031, Basel, Switzerland
| | - Andreas Beilhack
- Department of Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | - Sjoerd H van der Burg
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Albinusdreef 2, Leiden, 2333 ZA, The Netherlands
| | - Angela Riedel
- Mildred Scheel Early Career Center, University Hospital of Würzburg, Würzburg, Germany
| | - Benjamin Weide
- Department of Dermatology, University Medical Center Tübingen, Tübingen, Germany
| | | | - Jörg Wischhusen
- Department of Gynecology, University Hospital Würzburg, Würzburg, Germany.
| |
Collapse
|
66
|
Cui Y, Yuan T, Wang Y, Zheng D, Qin L, Li S, Jiang Z, Lin S, Guo W, Wang Z, Liang Z, Li Y, Yao Y, Liu X, Tang Q, Tu HY, Zhang XC, Tang Z, Wong N, Zhang Z, Qin D, Thiery JP, Xu K, Li P. T lymphocytes expressing the switchable chimeric Fc receptor CD64 exhibit augmented persistence and antitumor activity. Cell Rep 2023; 42:112797. [PMID: 37436890 DOI: 10.1016/j.celrep.2023.112797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 04/29/2023] [Accepted: 06/26/2023] [Indexed: 07/14/2023] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy lacks persistent efficacy with "on-target, off-tumor" toxicities for treating solid tumors. Thus, an antibody-guided switchable CAR vector, the chimeric Fc receptor CD64 (CFR64), composed of a CD64 extracellular domain, is designed. T cells expressing CFR64 exert more robust cytotoxicity against cancer cells than CFR T cells with high-affinity CD16 variant (CD16v) or CD32A as their extracellular domains. CFR64 T cells also exhibit better long-term cytotoxicity and resistance to T cell exhaustion compared with conventional CAR T cells. With trastuzumab, the immunological synapse (IS) established by CFR64 is more stable with lower intensity induction of downstream signaling than anti-HER2 CAR T cells. Moreover, CFR64 T cells exhibit fused mitochondria in response to stimulation, while CARH2 T cells contain predominantly punctate mitochondria. These results show that CFR64 T cells may serve as a controllable engineered T cell therapy with prolonged persistence and long-term antitumor activity.
Collapse
Affiliation(s)
- Yuanbin Cui
- China-New Zealand Joint Laboratory on Biomedicine and Health, State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Tingjie Yuan
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, the Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Guangzhou Laboratory, Guangzhou, China
| | - Ying Wang
- Blood Disease Institution, Xuzhou Medical University, Xuzhou, Jiangsu, China; Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Diwei Zheng
- China-New Zealand Joint Laboratory on Biomedicine and Health, State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Le Qin
- China-New Zealand Joint Laboratory on Biomedicine and Health, State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Shanglin Li
- China-New Zealand Joint Laboratory on Biomedicine and Health, State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Zhiwu Jiang
- China-New Zealand Joint Laboratory on Biomedicine and Health, State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Shouheng Lin
- China-New Zealand Joint Laboratory on Biomedicine and Health, State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Wenjing Guo
- China-New Zealand Joint Laboratory on Biomedicine and Health, State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Zhi Wang
- China-New Zealand Joint Laboratory on Biomedicine and Health, State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Zhaoduan Liang
- China-New Zealand Joint Laboratory on Biomedicine and Health, State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China; T-cell Immunity Optimized Cure (TIOC) Therapeutics Limited, Hangzhou, China
| | - Yi Li
- T-cell Immunity Optimized Cure (TIOC) Therapeutics Limited, Hangzhou, China
| | - Yao Yao
- China-New Zealand Joint Laboratory on Biomedicine and Health, State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Xingguo Liu
- China-New Zealand Joint Laboratory on Biomedicine and Health, State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Qiannan Tang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hai-Yan Tu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xu-Chao Zhang
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Zhaoyang Tang
- Guangdong Zhaotai InVivo Biomedicine Co. Ltd., Guangzhou, China
| | - Nathalie Wong
- Department of Surgery of the Faculty of Medicine, the Chinese University of Hong Kong (CUHK), Hong Kong, China
| | - Zhenfeng Zhang
- Department of Radiology, Translational Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumor Microenvironment, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Dajiang Qin
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, the Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | | | - Kailin Xu
- Blood Disease Institution, Xuzhou Medical University, Xuzhou, Jiangsu, China; Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Peng Li
- China-New Zealand Joint Laboratory on Biomedicine and Health, State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China; Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, the Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Department of Surgery of the Faculty of Medicine, the Chinese University of Hong Kong (CUHK), Hong Kong, China; Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, China.
| |
Collapse
|
67
|
Chen H, Xu X, Hu W, Wu S, Xiao J, Wu P, Wang X, Han X, Zhang Y, Zhang Y, Jiang N, Liu W, Lou C, Chen W, Xu C, Lou J. Self-programmed dynamics of T cell receptor condensation. Proc Natl Acad Sci U S A 2023; 120:e2217301120. [PMID: 37399423 PMCID: PMC10334747 DOI: 10.1073/pnas.2217301120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 06/01/2023] [Indexed: 07/05/2023] Open
Abstract
A common event upon receptor-ligand engagement is the formation of receptor clusters on the cell surface, in which signaling molecules are specifically recruited or excluded to form signaling hubs to regulate cellular events. These clusters are often transient and can be disassembled to terminate signaling. Despite the general relevance of dynamic receptor clustering in cell signaling, the regulatory mechanism underlying the dynamics is still poorly understood. As a major antigen receptor in the immune system, T cell receptors (TCR) form spatiotemporally dynamic clusters to mediate robust yet temporal signaling to induce adaptive immune responses. Here we identify a phase separation mechanism controlling dynamic TCR clustering and signaling. The TCR signaling component CD3ε chain can condensate with Lck kinase through phase separation to form TCR signalosomes for active antigen signaling. Lck-mediated CD3ε phosphorylation, however, switched its binding preference to Csk, a functional suppressor of Lck, to cause the dissolvement of TCR signalosomes. Modulating TCR/Lck condensation by targeting CD3ε interactions with Lck or Csk directly affects T cell activation and function, highlighting the importance of the phase separation mechanism. The self-programmed condensation and dissolvement is thus a built-in mechanism of TCR signaling and might be relevant to other receptors.
Collapse
Affiliation(s)
- Hui Chen
- Key Laboratory of RNA Biology, Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing100101, China
- University of Chinese Academy of Sciences, Beijing100049, China
| | - Xinyi Xu
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai200031, China
| | - Wei Hu
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang310003, China
| | - Songfang Wu
- Key Laboratory of RNA Biology, Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing100101, China
| | - Jianhui Xiao
- Key Laboratory of RNA Biology, Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing100101, China
- University of Chinese Academy of Sciences, Beijing100049, China
| | - Peng Wu
- Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang310058, China
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, Zhejiang310012, China
| | - Xiaowen Wang
- Key Laboratory of RNA Biology, Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing100101, China
- University of Chinese Academy of Sciences, Beijing100049, China
| | - Xuling Han
- Key Laboratory of RNA Biology, Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing100101, China
- University of Chinese Academy of Sciences, Beijing100049, China
| | - Yanruo Zhang
- Key Laboratory of RNA Biology, Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing100101, China
| | - Yong Zhang
- Key Laboratory of RNA Biology, Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing100101, China
- University of Chinese Academy of Sciences, Beijing100049, China
| | - Ning Jiang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA19104
| | - Wanli Liu
- State Key Laboratory of Membrane Biology, Center for Life Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Institute for Immunology, School of Life Sciences, Tsinghua University, Beijing100084, China
| | - Changjie Lou
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang150001, China
| | - Wei Chen
- Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang310058, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Ministry of Education Frontier Science Center for Brain Science & Brain-machine Integration, State Key Laboratory for Modern Optical Instrumentation Key Laboratory for Biomedical Engineering of the Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, Zhejiang310012, China
- Zhejiang Laboratory for Systems and Precision Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang311121, China
| | - Chenqi Xu
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai200031, China
- School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang310024, China
| | - Jizhong Lou
- Key Laboratory of RNA Biology, Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing100101, China
- University of Chinese Academy of Sciences, Beijing100049, China
| |
Collapse
|
68
|
Yin S, Wang J, Chen L, Mao M, Rahma I, Geng Y, Huang R, Tong X, Liu Y, Wu C, Chen Y, Li J. Circulating Th2-biased T follicular helper cells impede antiviral humoral responses during chronic hepatitis B infection through upregulating CTLA4. Antiviral Res 2023:105665. [PMID: 37421985 DOI: 10.1016/j.antiviral.2023.105665] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 05/22/2023] [Accepted: 07/05/2023] [Indexed: 07/10/2023]
Abstract
Failure in curing chronic hepatitis B (CHB) caused by hepatitis B virus (HBV) can lead to functional impairment of B cells. Cytotoxic T-lymphocyte associated antigen 4 (CTLA4) regulates B cell and T follicular helper (Tfh) cell differentiation. In addition, Tfh cells play a critical role in helping B cells generate antibodies upon pathogen exposure. Here, we analyzed the global and HBsAg-specific B cells and circulating Tfh (cTfh) cells using samples from treatment-naïve and Peg-IFN-α-treated CHB patients and healthy subjects. Compared to healthy subjects, CTLA4 expression was significantly increased in cTfh cells, from CHB patients. The frequency of CTLA4+cTfh2 cells was negatively correlated with that of HBsAg-specific resting memory B cells. Importantly, inhibition of CTLA4 restored HBsAb secretion and promoted plasma cell differentiation. In addition, CTLA4+cTfh2 cells from CHB patients were ineffective in providing B cell help. Both expression of CTLA4 in cTfh and cTfh2 cells and ratios of CLTA4+cTfh and CTLA4+cTfh2 cells were significantly decreased in Peg-IFN-α-treated CHB patients who showed complete responses. Thus, our results highlighted that cTh2-biased T follicular helper cells could impede antiviral humoral responses during chronic HBV infection by upregulating CTLA4, suggesting that further optimizing potent Tfh cell responses may promote functional cure of CHB.
Collapse
Affiliation(s)
- Shengxia Yin
- Department of Infectious Diseases, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China; Institute of Viruses and Infectious Diseases, Nanjing University, Jiangsu, China
| | - Jian Wang
- Department of Infectious Diseases, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China; Institute of Viruses and Infectious Diseases, Nanjing University, Jiangsu, China
| | - Lin Chen
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Minxin Mao
- Department of Infectious Diseases, Nanjing Drum Tower Hospital, Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Issa Rahma
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yu Geng
- Department of Infectious Diseases, Nanjing Drum Tower Hospital, Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Rui Huang
- Department of Infectious Diseases, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China; Institute of Viruses and Infectious Diseases, Nanjing University, Jiangsu, China
| | - Xin Tong
- Department of Infectious Diseases, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China; Institute of Viruses and Infectious Diseases, Nanjing University, Jiangsu, China
| | - Yong Liu
- Institute of Viruses and Infectious Diseases, Nanjing University, Jiangsu, China; Department of Experimental Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Chao Wu
- Department of Infectious Diseases, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China; Institute of Viruses and Infectious Diseases, Nanjing University, Jiangsu, China.
| | - Yuxin Chen
- Institute of Viruses and Infectious Diseases, Nanjing University, Jiangsu, China; Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Jie Li
- Department of Infectious Diseases, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China; Institute of Viruses and Infectious Diseases, Nanjing University, Jiangsu, China.
| |
Collapse
|
69
|
Nishi W, Wakamatsu E, Machiyama H, Matsushima R, Saito K, Yoshida Y, Nishikawa T, Takehara T, Toyota H, Furuhata M, Nishijima H, Takeuchi A, Azuma M, Suzuki M, Yokosuka T. Evaluation of therapeutic PD-1 antibodies by an advanced single-molecule imaging system detecting human PD-1 microclusters. Nat Commun 2023; 14:3157. [PMID: 37280233 PMCID: PMC10244369 DOI: 10.1038/s41467-023-38512-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 05/04/2023] [Indexed: 06/08/2023] Open
Abstract
With recent advances in immune checkpoint inhibitors (ICIs), immunotherapy has become the standard treatment for various malignant tumors. Their indications and dosages have been determined empirically, taking individually conducted clinical trials into consideration, but without a standard method to evaluate them. Here we establish an advanced imaging system to visualize human PD-1 microclusters, in which a minimal T cell receptor (TCR) signaling unit co-localizes with the inhibitory co-receptor PD-1 in vitro. In these microclusters PD-1 dephosphorylates both the TCR/CD3 complex and its downstream signaling molecules via the recruitment of a phosphatase, SHP2, upon stimulation with the ligand hPD-L1. In this system, blocking antibodies for hPD-1-hPD-L1 binding inhibits hPD-1 microcluster formation, and each therapeutic antibody (pembrolizumab, nivolumab, durvalumab and atezolizumab) is characterized by a proprietary optimal concentration and combinatorial efficiency enhancement. We propose that our imaging system could digitally evaluate PD-1-mediated T cell suppression to evaluate their clinical usefulness and to develop the most suitable combinations among ICIs or between ICIs and conventional cancer treatments.
Collapse
Affiliation(s)
- Wataru Nishi
- Department of Thoracic Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
- Department of Immunology, Tokyo Medical University, Tokyo, 160-8402, Japan
| | - Ei Wakamatsu
- Department of Immunology, Tokyo Medical University, Tokyo, 160-8402, Japan
| | - Hiroaki Machiyama
- Department of Immunology, Tokyo Medical University, Tokyo, 160-8402, Japan
| | - Ryohei Matsushima
- Department of Thoracic Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
- Department of Immunology, Tokyo Medical University, Tokyo, 160-8402, Japan
| | - Kensho Saito
- Department of Immunology, Tokyo Medical University, Tokyo, 160-8402, Japan
- School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, 192-0392, Japan
| | - Yosuke Yoshida
- Department of Immunology, Tokyo Medical University, Tokyo, 160-8402, Japan
- Department of Nephrology, Tokyo Medical University, Tokyo, 160-8402, Japan
| | - Tetsushi Nishikawa
- Department of Immunology, Tokyo Medical University, Tokyo, 160-8402, Japan
- Department of Dermatology, Tokyo Medical University, Tokyo, 160-0023, Japan
| | - Tomohiro Takehara
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Hiroko Toyota
- Department of Immunology, Tokyo Medical University, Tokyo, 160-8402, Japan
| | - Masae Furuhata
- Department of Immunology, Tokyo Medical University, Tokyo, 160-8402, Japan
| | - Hitoshi Nishijima
- Department of Immunology, Tokyo Medical University, Tokyo, 160-8402, Japan
| | - Arata Takeuchi
- Department of Immunology, Tokyo Medical University, Tokyo, 160-8402, Japan
| | - Miyuki Azuma
- Department of Molecular Immunology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, 113-8549, Japan
| | - Makoto Suzuki
- Department of Thoracic Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Tadashi Yokosuka
- Department of Immunology, Tokyo Medical University, Tokyo, 160-8402, Japan.
| |
Collapse
|
70
|
Nicosia M, Lee J, Beavers A, Kish D, Farr GW, McGuirk PR, Pelletier MF, Lathia JD, Fairchild RL, Valujskikh A. Water channel aquaporin 4 is required for T cell receptor mediated lymphocyte activation. J Leukoc Biol 2023; 113:544-554. [PMID: 36805947 PMCID: PMC10848298 DOI: 10.1093/jleuko/qiad010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 12/16/2022] [Accepted: 01/16/2023] [Indexed: 02/04/2023] Open
Abstract
Aquaporins are a family of ubiquitously expressed transmembrane water channels implicated in a broad range of physiological functions. We have previously reported that aquaporin 4 (AQP4) is expressed on T cells and that treatment with a small molecule AQP4 inhibitor significantly delays T cell mediated heart allograft rejection. Using either genetic deletion or small molecule inhibitor, we show that AQP4 supports T cell receptor mediated activation of both mouse and human T cells. Intact AQP4 is required for optimal T cell receptor (TCR)-related signaling events, including nuclear translocation of transcription factors and phosphorylation of proximal TCR signaling molecules. AQP4 deficiency or inhibition impairs actin cytoskeleton rearrangements following TCR crosslinking, causing inferior TCR polarization and a loss of TCR signaling. Our findings reveal a novel function of AQP4 in T lymphocytes and identify AQP4 as a potential therapeutic target for preventing TCR-mediated T cell activation.
Collapse
Affiliation(s)
- Michael Nicosia
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Juyeun Lee
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Ashley Beavers
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Danielle Kish
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - George W. Farr
- Aeromics Inc., 470 James Street Suite 007, New Haven, CT 06513, United States
| | - Paul R. McGuirk
- Aeromics Inc., 470 James Street Suite 007, New Haven, CT 06513, United States
| | - Marc F. Pelletier
- Aeromics Inc., 470 James Street Suite 007, New Haven, CT 06513, United States
| | - Justin D. Lathia
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Robert L. Fairchild
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Anna Valujskikh
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| |
Collapse
|
71
|
Jürgens C, Ssebyatika G, Beyer S, Plückebaum N, Kropp KA, González-Motos V, Ritter B, Böning H, Nikolouli E, Kinchington PR, Lachmann N, Depledge DP, Krey T, Viejo-Borbolla A. Viral modulation of type II interferon increases T cell adhesion and virus spread. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.26.542397. [PMID: 37292914 PMCID: PMC10246016 DOI: 10.1101/2023.05.26.542397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
During primary infection, varicella zoster virus (VZV) infects epithelial cells in the respiratory lymphoid organs and mucosa. Subsequent infection of lymphocytes, T cells in particular, causes primary viremia allowing systemic spread throughout the host, including the skin. This results in the expression of cytokines, including interferons (IFNs) which partly limit primary infection. VZV also spreads from skin keratinocytes to lymphocytes prior to secondary viremia. How VZV infects lymphocytes from epithelial cells while evading the cytokine response has not been fully established. Here, we show that VZV glycoprotein C (gC) binds IFN-γ and modifies its activity. Transcriptomic analysis revealed that gC in combination with IFN-γ increased the expression of a small subset of IFN-stimulated genes (ISGs), including intercellular adhesion molecule 1 (ICAM1), as well as several chemokines and immunomodulatory genes. The higher ICAM1 protein level at the plasma membrane of epithelial cells resulted in lymphocyte function-associated antigen 1 (LFA-1)-dependent T cell adhesion. This gC activity required a stable interaction with IFN-γ and signalling through the IFN-γ receptor. Finally, the presence of gC during infection increased VZV spread from epithelial cells to peripheral blood mononuclear cells. This constitutes the discovery of a novel strategy to modulate the activity of IFN-γ, inducing the expression of a subset of ISGs, leading to enhanced T cell adhesion and virus spread.
Collapse
Affiliation(s)
- Carina Jürgens
- Institute of Virology, Hannover Medical School, Hannover 30625, Germany
| | - George Ssebyatika
- Institute of Virology, Hannover Medical School, Hannover 30625, Germany
- Institute of Biochemistry, University of Lübeck, Lübeck 23562, Germany
| | - Sarah Beyer
- Institute of Virology, Hannover Medical School, Hannover 30625, Germany
| | - Nina Plückebaum
- Institute of Virology, Hannover Medical School, Hannover 30625, Germany
| | - Kai A. Kropp
- Institute of Virology, Hannover Medical School, Hannover 30625, Germany
| | - Víctor González-Motos
- Institute of Virology, Hannover Medical School, Hannover 30625, Germany
- University of Veterinary Medicine Hannover, Foundation, Hannover 30559, Germany
| | - Birgit Ritter
- Institute of Virology, Hannover Medical School, Hannover 30625, Germany
| | - Heike Böning
- Institute of Virology, Hannover Medical School, Hannover 30625, Germany
| | - Eirini Nikolouli
- Department for Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover 30625, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Nikolai-Fuchs-Str. 1, 30625 Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover, Germany
- Excellence Cluster 2155 RESIST, Hannover Medical School, Hannover 30625, Germany
| | - Paul R. Kinchington
- Department of Ophthalmology and of Molecular Microbiology and Genetics, University of Pittsburgh, Pittsburgh, PA, United States
| | - Nico Lachmann
- Department for Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover 30625, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Nikolai-Fuchs-Str. 1, 30625 Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover, Germany
- Excellence Cluster 2155 RESIST, Hannover Medical School, Hannover 30625, Germany
| | - Daniel Pearce Depledge
- Institute of Virology, Hannover Medical School, Hannover 30625, Germany
- Excellence Cluster 2155 RESIST, Hannover Medical School, Hannover 30625, Germany
- German Center for Infection Research (DZIF), partner site Hannover-Braunschweig, Hannover, Germany
| | - Thomas Krey
- Institute of Virology, Hannover Medical School, Hannover 30625, Germany
- Institute of Biochemistry, University of Lübeck, Lübeck 23562, Germany
- Excellence Cluster 2155 RESIST, Hannover Medical School, Hannover 30625, Germany
- Centre for Structural Systems Biology (CSSB), 22607 Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, 22607 Hamburg, Germany
| | - Abel Viejo-Borbolla
- Institute of Virology, Hannover Medical School, Hannover 30625, Germany
- Excellence Cluster 2155 RESIST, Hannover Medical School, Hannover 30625, Germany
| |
Collapse
|
72
|
Haydinger CD, Ashander LM, Tan ACR, Smith JR. Intercellular Adhesion Molecule 1: More than a Leukocyte Adhesion Molecule. BIOLOGY 2023; 12:biology12050743. [PMID: 37237555 DOI: 10.3390/biology12050743] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/15/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023]
Abstract
Intercellular adhesion molecule 1 (ICAM-1) is a transmembrane protein in the immunoglobulin superfamily expressed on the surface of multiple cell populations and upregulated by inflammatory stimuli. It mediates cellular adhesive interactions by binding to the β2 integrins macrophage antigen 1 and leukocyte function-associated antigen 1, as well as other ligands. It has important roles in the immune system, including in leukocyte adhesion to the endothelium and transendothelial migration, and at the immunological synapse formed between lymphocytes and antigen-presenting cells. ICAM-1 has also been implicated in the pathophysiology of diverse diseases from cardiovascular diseases to autoimmune disorders, certain infections, and cancer. In this review, we summarize the current understanding of the structure and regulation of the ICAM1 gene and the ICAM-1 protein. We discuss the roles of ICAM-1 in the normal immune system and a selection of diseases to highlight the breadth and often double-edged nature of its functions. Finally, we discuss current therapeutics and opportunities for advancements.
Collapse
Affiliation(s)
- Cameron D Haydinger
- College of Medicine and Public Health, Flinders University, Adelaide, SA 5042, Australia
| | - Liam M Ashander
- College of Medicine and Public Health, Flinders University, Adelaide, SA 5042, Australia
| | - Alwin Chun Rong Tan
- College of Medicine and Public Health, Flinders University, Adelaide, SA 5042, Australia
| | - Justine R Smith
- College of Medicine and Public Health, Flinders University, Adelaide, SA 5042, Australia
| |
Collapse
|
73
|
Tabaei SR, Fernandez-Villamarin M, Vafaei S, Rooney L, Mendes PM. Recapitulating the Lateral Organization of Membrane Receptors at the Nanoscale. ACS NANO 2023. [PMID: 37200265 DOI: 10.1021/acsnano.3c00683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Many cell membrane functions emerge from the lateral presentation of membrane receptors. The link between the nanoscale organization of the receptors and ligand binding remains, however, mostly unclear. In this work, we applied surface molecular imprinting and utilized the phase behavior of lipid bilayers to create platforms that recapitulate the lateral organization of membrane receptors at the nanoscale. We used liposomes decorated with amphiphilic boronic acids that commonly serve as synthetic saccharide receptors and generated three lateral modes of receptor presentation─random distribution, nanoclustering, and receptor crowding─and studied their interaction with saccharides. In comparison to liposomes with randomly dispersed receptors, surface-imprinted liposomes resulted in more than a 5-fold increase in avidity. Quantifying the binding affinity and cooperativity proved that the boost was mediated by the formation of the nanoclusters rather than a local increase in the receptor concentration. In contrast, receptor crowding, despite the presence of increased local receptor concentrations, prevented multivalent oligosaccharide binding due to steric effects. The findings demonstrate the significance of nanometric aspects of receptor presentation and generation of multivalent ligands including artificial lectins for the sensitive and specific detection of glycans.
Collapse
Affiliation(s)
- Seyed R Tabaei
- School of Chemistry and Chemical Engineering, Queen's University Belfast, Stranmillis Road, Belfast, BT9 5AG, U.K
| | | | - Setareh Vafaei
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B15 2TT, U.K
| | - Lorcan Rooney
- School of Chemistry and Chemical Engineering, Queen's University Belfast, Stranmillis Road, Belfast, BT9 5AG, U.K
| | - Paula M Mendes
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B15 2TT, U.K
| |
Collapse
|
74
|
Chang HF, Schirra C, Pattu V, Krause E, Becherer U. Lytic granule exocytosis at immune synapses: lessons from neuronal synapses. Front Immunol 2023; 14:1177670. [PMID: 37275872 PMCID: PMC10233144 DOI: 10.3389/fimmu.2023.1177670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 05/09/2023] [Indexed: 06/07/2023] Open
Abstract
Regulated exocytosis is a central mechanism of cellular communication. It is not only the basis for neurotransmission and hormone release, but also plays an important role in the immune system for the release of cytokines and cytotoxic molecules. In cytotoxic T lymphocytes (CTLs), the formation of the immunological synapse is required for the delivery of the cytotoxic substances such as granzymes and perforin, which are stored in lytic granules and released via exocytosis. The molecular mechanisms of their fusion with the plasma membrane are only partially understood. In this review, we discuss the molecular players involved in the regulated exocytosis of CTL, highlighting the parallels and differences to neuronal synaptic transmission. Additionally, we examine the strengths and weaknesses of both systems to study exocytosis.
Collapse
|
75
|
Sadhu L, Tsopoulidis N, Hasanuzzaman M, Laketa V, Way M, Fackler OT. ARPC5 isoforms and their regulation by calcium-calmodulin-N-WASP drive distinct Arp2/3-dependent actin remodeling events in CD4 T cells. eLife 2023; 12:e82450. [PMID: 37162507 PMCID: PMC10171864 DOI: 10.7554/elife.82450] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 04/06/2023] [Indexed: 05/11/2023] Open
Abstract
CD4 T cell activation induces nuclear and cytoplasmic actin polymerization via the Arp2/3 complex to activate cytokine expression and strengthen T cell receptor (TCR) signaling. Actin polymerization dynamics and filament morphology differ between nucleus and cytoplasm. However, it is unclear how the Arp2/3 complex mediates distinct nuclear and cytoplasmic actin polymerization in response to a common stimulus. In humans, the ARP3, ARPC1, and ARPC5 subunits of the Arp2/3 complex exist as two different isoforms, resulting in complexes with different properties. Here, we show that the Arp2/3 subunit isoforms ARPC5 and ARPC5L play a central role in coordinating distinct actin polymerization events in CD4 T cells. While ARPC5L is heterogeneously expressed in individual CD4 T cells, it specifically drives nuclear actin polymerization upon T cell activation. In contrast, ARPC5 is evenly expressed in CD4 T cell populations and is required for cytoplasmic actin dynamics. Interestingly, nuclear actin polymerization triggered by a different stimulus, DNA replication stress, specifically requires ARPC5 but not ARPC5L. TCR signaling but not DNA replication stress induces nuclear actin polymerization via nuclear calcium-calmodulin signaling and N-WASP. Diversity in the molecular properties and individual expression patterns of ARPC5 subunit isoforms thus tailors Arp2/3-mediated actin polymerization to different physiological stimuli.
Collapse
Affiliation(s)
- Lopamudra Sadhu
- Department of Infectious Diseases, Integrative Virology, University Hospital HeidelbergHeidelbergGermany
| | - Nikolaos Tsopoulidis
- Department of Infectious Diseases, Integrative Virology, University Hospital HeidelbergHeidelbergGermany
| | - Md Hasanuzzaman
- Department of Infectious Diseases, Integrative Virology, University Hospital HeidelbergHeidelbergGermany
| | - Vibor Laketa
- Department of Infectious Diseases, Virology, University Hospital HeidelbergHeidelbergGermany
| | - Michael Way
- Cellular Signalling and Cytoskeletal Function Laboratory, The Francis Crick InstituteLondonUnited Kingdom
- Department of Infectious Disease, Imperial CollegeLondonUnited Kingdom
| | - Oliver T Fackler
- Department of Infectious Diseases, Integrative Virology, University Hospital HeidelbergHeidelbergGermany
| |
Collapse
|
76
|
Ravindran S. QnAs with Michael L. Dustin. Proc Natl Acad Sci U S A 2023; 120:e2304200120. [PMID: 37040410 PMCID: PMC10120066 DOI: 10.1073/pnas.2304200120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2023] Open
|
77
|
Shi H, Shao B. LFA-1 Activation in T-Cell Migration and Immunological Synapse Formation. Cells 2023; 12:cells12081136. [PMID: 37190045 DOI: 10.3390/cells12081136] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/02/2023] [Accepted: 04/06/2023] [Indexed: 05/17/2023] Open
Abstract
Integrin LFA-1 plays a critical role in T-cell migration and in the formation of immunological synapses. LFA-1 functions through interacting with its ligands with differing affinities: low, intermediate, and high. Most prior research has studied how LFA-1 in the high-affinity state regulates the trafficking and functions of T cells. LFA-1 is also presented in the intermediate-affinity state on T cells, however, the signaling to activate LFA-1 to the intermediate-affinity state and the role of LFA-1 in this affinity state both remain largely elusive. This review briefly summarizes the activation and roles of LFA-1 with varied ligand-binding affinities in the regulation of T-cell migration and immunological synapse formation.
Collapse
Affiliation(s)
- Huiping Shi
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Bojing Shao
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| |
Collapse
|
78
|
Basu I, Li H, Trease AJ, Sorgen PL. Regulation of Cx43 Gap Junction Intercellular Communication by Bruton's Tyrosine Kinase and Interleukin-2-Inducible T-Cell Kinase. Biomolecules 2023; 13:biom13040660. [PMID: 37189407 DOI: 10.3390/biom13040660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/30/2023] [Accepted: 04/06/2023] [Indexed: 05/17/2023] Open
Abstract
T and B cell receptor signaling involves the activation of Akt, MAPKs, and PKC as well as an increase in intracellular Ca2+ and calmodulin activation. While these coordinate the rapid turnover of gap junctions, also implicated in this process is Src, which is not activated as part of T and B cell receptor signaling. An in vitro kinase screen identified that Bruton's tyrosine kinase (BTK) and interleukin-2-inducible T-cell kinase (ITK) phosphorylate Cx43. Mass spectroscopy revealed that BTK and ITK phosphorylate Cx43 residues Y247, Y265, and Y313, which are identical to the residues phosphorylated by Src. Overexpression of BTK or ITK in the HEK-293T cells led to increased Cx43 tyrosine phosphorylation as well as decreased gap junction intercellular communication (GJIC) and Cx43 membrane localization. In the lymphocytes, activation of the B cell receptor (Daudi cells) or T cell receptor (Jurkat cells) increased the BTK and ITK activity, respectively. While this led to increased tyrosine phosphorylation of Cx43 and decreased GJIC, the cellular localization of Cx43 changed little. We have previously identified that Pyk2 and Tyk2 also phosphorylate Cx43 at residues Y247, Y265, and Y313 with a similar cellular fate to that of Src. With phosphorylation critical to Cx43 assembly and turnover, and kinase expression varying between different cell types, there would be a need for different kinases to achieve the same regulation of Cx43. The work presented herein suggests that in the immune system, ITK and BTK have the capacity for the tyrosine phosphorylation of Cx43 to alter the gap junction function in a similar manner as Pyk2, Tyk2, and Src.
Collapse
Affiliation(s)
- Ishika Basu
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Hanjun Li
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Andrew J Trease
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Paul L Sorgen
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
79
|
Jenkins E, Körbel M, O'Brien-Ball C, McColl J, Chen KY, Kotowski M, Humphrey J, Lippert AH, Brouwer H, Santos AM, Lee SF, Davis SJ, Klenerman D. Antigen discrimination by T cells relies on size-constrained microvillar contact. Nat Commun 2023; 14:1611. [PMID: 36959206 PMCID: PMC10036606 DOI: 10.1038/s41467-023-36855-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 02/21/2023] [Indexed: 03/25/2023] Open
Abstract
T cells use finger-like protrusions called 'microvilli' to interrogate their targets, but why they do so is unknown. To form contacts, T cells must overcome the highly charged, barrier-like layer of large molecules forming a target cell's glycocalyx. Here, T cells are observed to use microvilli to breach a model glycocalyx barrier, forming numerous small (<0.5 μm diameter) contacts each of which is stabilized by the small adhesive protein CD2 expressed by the T cell, and excludes large proteins including CD45, allowing sensitive, antigen dependent TCR signaling. In the absence of the glycocalyx or when microvillar contact-size is increased by enhancing CD2 expression, strong signaling occurs that is no longer antigen dependent. Our observations suggest that, modulated by the opposing effects of the target cell glycocalyx and small adhesive proteins, the use of microvilli equips T cells with the ability to effect discriminatory receptor signaling.
Collapse
Affiliation(s)
- Edward Jenkins
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
- Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
| | - Markus Körbel
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Caitlin O'Brien-Ball
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
- Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
| | - James McColl
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Kevin Y Chen
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Mateusz Kotowski
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
- Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
| | - Jane Humphrey
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Anna H Lippert
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Heather Brouwer
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
- Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
| | - Ana Mafalda Santos
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
- Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
| | - Steven F Lee
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Simon J Davis
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK.
- Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK.
| | - David Klenerman
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK.
| |
Collapse
|
80
|
Ide H, Aoshi T, Saito M, Espulgar WV, Briones JC, Hosokawa M, Matsunaga H, Arikawa K, Takeyama H, Koyama S, Takamatsu H, Tamiya E. Linking antigen specific T-cell dynamics in a microfluidic chip to single cell transcription patterns. Biochem Biophys Res Commun 2023; 657:8-15. [PMID: 36963175 DOI: 10.1016/j.bbrc.2023.03.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/10/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023]
Abstract
A new non-invasive screening profile has been realized that can aid in determining T-cell activation state at single-cell level. Production of activated T-cells with good specificity and stable proliferation is greatly beneficial for advancing adoptive immunotherapy as innate immunological cells are not effective in recognizing and eliminating cancer as expected. The screening method is realized by relating intracellular Ca2+ intensity and motility of T-cells interacting with APC (Antigen Presenting Cells) in a microfluidic chip. The system is tested using APC pulsed with OVA257-264 peptide and its modified affinities (N4, Q4, T4 and V4), and the T-cells from OT-1 mice. In addition, single cell RNA sequencing reveals the activation states of the cells and the clusters from the derived profiles can be indicative of the T-cell activation state. The presented system here can be versatile for a comprehensive application to proceed with T-cell-based immunotherapy and screen the antigen-specific T-cells with excellent efficiency and high proliferation.
Collapse
Affiliation(s)
- Hiroki Ide
- Graduate School of Engineering Osaka Univ, Japan; PhotoBIO Lab, AIST-Osaka Univ, Japan
| | - Taiki Aoshi
- Research Institute for Microbial Diseases, Osaka Univ, Japan
| | - Masato Saito
- PhotoBIO Lab, AIST-Osaka Univ, Japan; Life and Medical Photonics Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan.
| | | | - Jonathan Campos Briones
- Life and Medical Photonics Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan
| | - Masahito Hosokawa
- Department of Life Science and Medical Bioscience, Waseda Univ, Japan; CBBD-OIL, AIST-Waseda Univ, Japan; Institute for Advanced Research of Biosystem Dynamics, Waseda Research Institute for Science and Engineering, Waseda Univ, Japan; Research Organization for Nano and Life Innovation, Waseda Univ, Japan
| | - Hiroko Matsunaga
- Research Organization for Nano and Life Innovation, Waseda Univ, Japan
| | - Koji Arikawa
- Research Organization for Nano and Life Innovation, Waseda Univ, Japan
| | - Haruko Takeyama
- Department of Life Science and Medical Bioscience, Waseda Univ, Japan; CBBD-OIL, AIST-Waseda Univ, Japan; Institute for Advanced Research of Biosystem Dynamics, Waseda Research Institute for Science and Engineering, Waseda Univ, Japan; Research Organization for Nano and Life Innovation, Waseda Univ, Japan
| | | | | | - Eiichi Tamiya
- PhotoBIO Lab, AIST-Osaka Univ, Japan; Institute of Scientific and Industrial Research, Osaka University, Japan
| |
Collapse
|
81
|
Anto NP, Muraleedharan A, Nath PR, Sun Z, Keasar C, Livneh E, Braiman A, Altman A, Kong KF, Isakov N. The Peptidyl-Prolyl cis-trans isomerase, Pin1, associates with Protein Kinase C θ via a critical Phospho-Thr-Pro motif in the V3 regulatory domain. Front Immunol 2023; 14:1126464. [PMID: 36969236 PMCID: PMC10031136 DOI: 10.3389/fimmu.2023.1126464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 02/27/2023] [Indexed: 03/11/2023] Open
Abstract
Protein kinase C-θ (PKCθ) is a member of the novel PKC subfamily known for its selective and predominant expression in T lymphocytes where it regulates essential functions required for T cell activation and proliferation. Our previous studies provided a mechanistic explanation for the recruitment of PKCθ to the center of the immunological synapse (IS) by demonstrating that a proline-rich (PR) motif within the V3 region in the regulatory domain of PKCθ is necessary and sufficient for PKCθ IS localization and function. Herein, we highlight the importance of Thr335-Pro residue in the PR motif, the phosphorylation of which is key in the activation of PKCθ and its subsequent IS localization. We demonstrate that the phospho-Thr335-Pro motif serves as a putative binding site for the peptidyl-prolyl cis-trans isomerase (PPIase), Pin1, an enzyme that specifically recognizes peptide bonds at phospho-Ser/Thr-Pro motifs. Binding assays revealed that mutagenesis of PKCθ-Thr335-to-Ala abolished the ability of PKCθ to interact with Pin1, while Thr335 replacement by a Glu phosphomimetic, restored PKCθ binding to Pin1, suggesting that Pin1-PKCθ association is contingent upon the phosphorylation of the PKCθ-Thr335-Pro motif. Similarly, the Pin1 mutant, R17A, failed to associate with PKCθ, suggesting that the integrity of the Pin1 N-terminal WW domain is a requisite for Pin1-PKCθ interaction. In silico docking studies underpinned the role of critical residues in the Pin1-WW domain and the PKCθ phospho-Thr335-Pro motif, to form a stable interaction between Pin1 and PKCθ. Furthermore, TCR crosslinking in human Jurkat T cells and C57BL/6J mouse-derived splenic T cells promoted a rapid and transient formation of Pin1-PKCθ complexes, which followed a T cell activation-dependent temporal kinetic, suggesting a role for Pin1 in PKCθ-dependent early activation events in TCR-triggered T cells. PPIases that belong to other subfamilies, i.e., cyclophilin A or FK506-binding protein, failed to associate with PKCθ, indicating the specificity of the Pin1-PKCθ association. Fluorescent cell staining and imaging analyses demonstrated that TCR/CD3 triggering promotes the colocalization of PKCθ and Pin1 at the cell membrane. Furthermore, interaction of influenza hemagglutinin peptide (HA307-319)-specific T cells with antigen-fed antigen presenting cells (APCs) led to colocalization of PKCθ and Pin1 at the center of the IS. Together, we point to an uncovered function for the Thr335-Pro motif within the PKCθ-V3 regulatory domain to serve as a priming site for its activation upon phosphorylation and highlight its tenability to serve as a regulatory site for the Pin1 cis-trans isomerase.
Collapse
Affiliation(s)
- Nikhil Ponnoor Anto
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Amitha Muraleedharan
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Pulak Ranjan Nath
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Zuoming Sun
- Department of Immunology and Theranostics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Chen Keasar
- The Department of Computer Science, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Etta Livneh
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Alex Braiman
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Amnon Altman
- Division of Cell Biology, La Jolla Institute for Immunology, San Diego, CA, United States
| | - Kok-Fai Kong
- Division of Cell Biology, La Jolla Institute for Immunology, San Diego, CA, United States
| | - Noah Isakov
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
- *Correspondence: Noah Isakov,
| |
Collapse
|
82
|
Beppler C, Eichorst J, Marchuk K, Cai E, Castellanos CA, Sriram V, Roybal KT, Krummel MF. Hyperstabilization of T cell microvilli contacts by chimeric antigen receptors. J Cell Biol 2023; 222:e202205118. [PMID: 36520493 PMCID: PMC9757849 DOI: 10.1083/jcb.202205118] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 10/25/2022] [Accepted: 11/29/2022] [Indexed: 12/23/2022] Open
Abstract
T cells typically recognize their ligands using a defined cell biology-the scanning of their membrane microvilli (MV) to palpate their environment-while that same membrane scaffolds T cell receptors (TCRs) that can signal upon ligand binding. Chimeric antigen receptors (CARs) present both a therapeutic promise and a tractable means to study the interplay between receptor affinity, MV dynamics and T cell function. CARs are often built using single-chain variable fragments (scFvs) with far greater affinity than that of natural TCRs. We used high-resolution lattice lightsheet (LLS) and total internal reflection fluorescence (TIRF) imaging to visualize MV scanning in the context of variations in CAR design. This demonstrated that conventional CARs hyper-stabilized microvillar contacts relative to TCRs. Reducing receptor affinity, antigen density, and/or multiplicity of receptor binding sites normalized microvillar dynamics and synapse resolution, and effector functions improved with reduced affinity and/or antigen density, highlighting the importance of understanding the underlying cell biology when designing receptors for optimal antigen engagement.
Collapse
Affiliation(s)
- Casey Beppler
- Department of Pathology and ImmunoX, University of California, San Francisco, San Francisco, CA, USA
| | - John Eichorst
- Biological Imaging Development CoLab, University of California, San Francisco, San Francisco, CA, USA
| | - Kyle Marchuk
- Biological Imaging Development CoLab, University of California, San Francisco, San Francisco, CA, USA
| | - En Cai
- Department of Pathology and ImmunoX, University of California, San Francisco, San Francisco, CA, USA
| | - Carlos A. Castellanos
- Department of Microbiology and Immunology, Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA, USA
| | | | - Kole T. Roybal
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
- Helen Diller Comprehensive Cancer Center, San Francisco, CA, USA
| | - Matthew F. Krummel
- Department of Pathology and ImmunoX, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
83
|
White D, Cote-Martin A, Bleck M, Garaffa N, Shaaban A, Wu H, Liu D, Young D, Scheer J, Lorenz IC, Nixon A, Fine JS, Byrne FR, Mbow ML, Moreno-Garcia ME. Programmed Cell Death-1 (PD-1) anchoring to the GPI-linked co-receptor CD48 reveals a novel mechanism to modulate PD-1-dependent inhibition of human T cells. Mol Immunol 2023; 156:31-38. [PMID: 36889184 DOI: 10.1016/j.molimm.2023.02.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 02/16/2023] [Indexed: 03/08/2023]
Abstract
Activation of PD-1 by anchoring it to Antigen Receptor (AR) components or associated co-receptors represents an attractive approach to treat autoimmune conditions. In this study, we provide evidence that CD48, a common lipid raft and Src kinase-associated coreceptor, induces significant Src kinase-dependent activation of PD-1 upon crosslinking, while CD71, a receptor excluded from these compartments, does not. Functionally, using bead-conjugated antibodies we demonstrate that CD48-dependent activation of PD-1 inhibits proliferation of AR-induced primary human T cells, and similarly, PD-1 activation using PD-1/CD48 bispecific antibodies inhibits IL-2, enhances IL-10 secretion, and reduces NFAT activation in primary human and Jurkat T cells, respectively. As a whole, CD48-dependent activation of PD-1 represents a novel mechanism to fine tune T cell activation, and by functionally anchoring PD-1 with receptors other than AR, this study provides a conceptual framework for rational development of novel therapies that activate inhibitory checkpoint receptors for treatment of immune-mediated diseases.
Collapse
Affiliation(s)
- Della White
- Departments of Immunology and Respiratory Research, USA.
| | | | - Marina Bleck
- Departments of Immunology and Respiratory Research, USA
| | | | | | - Helen Wu
- Biotherapeutics Discovery. Boehringer Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, CT 06811 USA
| | - Dongmei Liu
- Biotherapeutics Discovery. Boehringer Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, CT 06811 USA
| | - David Young
- Biotherapeutics Discovery. Boehringer Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, CT 06811 USA
| | - Justin Scheer
- Biotherapeutics Discovery. Boehringer Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, CT 06811 USA
| | - Ivo C Lorenz
- Biotherapeutics Discovery. Boehringer Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, CT 06811 USA
| | - Andrew Nixon
- Biotherapeutics Discovery. Boehringer Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, CT 06811 USA
| | - Jay S Fine
- Departments of Immunology and Respiratory Research, USA
| | | | - M Lamine Mbow
- Departments of Immunology and Respiratory Research, USA; Cancer Immunology and Immune Modulation, USA
| | | |
Collapse
|
84
|
Chien CY, Lin JC, Huang CY, Hsu CY, Yang KC, Chattopadhyay S, Nikoloutsos N, Hsieh PCH, Hu CMJ. In Situ Hydrogelation of Cellular Monolayers Enables Conformal Biomembrane Functionalization for Xeno-Free Feeder Substrate Engineering. Adv Healthc Mater 2023; 12:e2201708. [PMID: 36455286 DOI: 10.1002/adhm.202201708] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 11/14/2022] [Indexed: 12/03/2022]
Abstract
The intricate functionalities of cellular membranes have inspired strategies for deriving and anchoring cell-surface components onto solid substrates for biological studies, biosensor applications, and tissue engineering. However, introducing conformal and right-side-out cell membrane coverage onto planar substrates requires cumbersome protocols susceptible to significant device-to-device variability. Here, a facile approach for biomembrane functionalization of planar substrates is demonstrated by subjecting confluent cellular monolayer to intracellular hydrogel polymerization. The resulting cell-gel hybrid, herein termed GELL (gelated cell), exhibits extraordinary stability and retains the structural integrity, membrane fluidity, membrane protein mobility, and topology of living cells. In assessing the utility of GELL layers as a tissue engineering feeder substrate for stem cell maintenance, GELL feeder prepared from primary mouse embryonic fibroblasts not only preserves the stemness of murine stem cells but also exhibits advantages over live feeder cells owing to the GELL's inanimate, non-metabolizing nature. The preparation of a xeno-free feeder substrate devoid of non-human components is further shown with HeLa cells, and the resulting HeLa GELL feeder effectively sustains the growth and stemness of both murine and human induced pluripotent stem cells. The study highlights a novel bio-functionalization strategy that introduces new opportunities for tissue engineering and other biomedical applications.
Collapse
Affiliation(s)
- Chen-Ying Chien
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan
| | - Jung-Chen Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan
- Institute of Oral Biology, College of Dentistry, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
| | - Ching-Ying Huang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan
| | - Chung-Yao Hsu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academic Sinica, Taipei, 11529, Taiwan
| | - Kai-Chieh Yang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academic Sinica, Taipei, 11529, Taiwan
| | - Saborni Chattopadhyay
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academic Sinica, Taipei, 11529, Taiwan
| | | | | | - Che-Ming Jack Hu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academic Sinica, Taipei, 11529, Taiwan
- Center of Applied Nanomedicine, National Cheng Kung University, Tainan, 701, Taiwan
| |
Collapse
|
85
|
Chew PY, Joseph JA, Collepardo-Guevara R, Reinhardt A. Thermodynamic origins of two-component multiphase condensates of proteins. Chem Sci 2023; 14:1820-1836. [PMID: 36819870 PMCID: PMC9931050 DOI: 10.1039/d2sc05873a] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 01/06/2023] [Indexed: 01/26/2023] Open
Abstract
Intracellular condensates are highly multi-component systems in which complex phase behaviour can ensue, including the formation of architectures comprising multiple immiscible condensed phases. Relying solely on physical intuition to manipulate such condensates is difficult because of the complexity of their composition, and systematically learning the underlying rules experimentally would be extremely costly. We address this challenge by developing a computational approach to design pairs of protein sequences that result in well-separated multilayered condensates and elucidate the molecular origins of these compartments. Our method couples a genetic algorithm to a residue-resolution coarse-grained protein model. We demonstrate that we can design protein partners to form multiphase condensates containing naturally occurring proteins, such as the low-complexity domain of hnRNPA1 and its mutants, and show how homo- and heterotypic interactions must differ between proteins to result in multiphasicity. We also show that in some cases the specific pattern of amino-acid residues plays an important role. Our findings have wide-ranging implications for understanding and controlling the organisation, functions and material properties of biomolecular condensates.
Collapse
Affiliation(s)
- Pin Yu Chew
- Yusuf Hamied Department of Chemistry, University of Cambridge Cambridge CB2 1EW UK
| | - Jerelle A Joseph
- Yusuf Hamied Department of Chemistry, University of Cambridge Cambridge CB2 1EW UK
- Department of Physics, University of Cambridge Cambridge CB3 0HE UK
- Department of Genetics, University of Cambridge Cambridge CB2 3EH UK
| | - Rosana Collepardo-Guevara
- Yusuf Hamied Department of Chemistry, University of Cambridge Cambridge CB2 1EW UK
- Department of Physics, University of Cambridge Cambridge CB3 0HE UK
- Department of Genetics, University of Cambridge Cambridge CB2 3EH UK
| | - Aleks Reinhardt
- Yusuf Hamied Department of Chemistry, University of Cambridge Cambridge CB2 1EW UK
| |
Collapse
|
86
|
Li K, Chen Y, Zhu N, Chen S, Jia M, Xue L, Hao M, Zhang C. Real-time detection of T cell activation by visualizing TCR nanoclusters with a cholesterol derived aggregation-induced emission probe. Eur J Med Chem 2023; 247:115073. [PMID: 36603511 DOI: 10.1016/j.ejmech.2022.115073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 12/18/2022] [Accepted: 12/28/2022] [Indexed: 12/30/2022]
Abstract
Successful T-cell based immunotherapy usually depends on the activation of T cells. Most of commonly used methods for assessing T cell activity rely on the antibody-based technology, which focus on detecting protein-centered activation markers, including CD25, cytokines and so on. However, these methods always involve tedious sample-preparation process, labor-consuming and costly, which could not be utilized in real-time detection. The T cell receptor (TCR) clustering is another kind of essential T cell activation marker on the membrane, which increases during the activation state of T cells. We herein developed a cholesterol derived aggregation-induced emission (AIE) fluorescent probe (R-TPE-PEG-Chol) for detecting T cell activation in real-time. Five probes were first designed and synthesized and among them COOH-TPE-PEG-Chol displayed the best imaging effects, which had no significant impact on the key physiological functions of T cells. In addition, we have proved that COOH-TPE-PEG-Chol was introduced onto the naïve T cell membrane in its molecularly dissolved form without fluorescent emission. While during T cell activation, the formation of TCR nanoclusters would induce aggregation of membrane cholesterol, which could provoke the fluorescence signal of the COOH-TPE-PEG-Chol due to the AIE characteristic. Moreover, the enhancement of the fluorescence intensity was positively related to the activation state of T cells. Our study demonstrated the concept of cholesterol-derived AIE fluorescent probes for deciphering the spatiotemporal arrangements of TCR on the membrane during T cell activation, and consequently provided a novel and complementary strategy for detecting T cell activation in real-time.
Collapse
Affiliation(s)
- Kaiming Li
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing, 210009, China; Chongqing Innovation Institute of China Pharmaceutical University, Chongqing, 401135, China
| | - Yue Chen
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing, 210009, China
| | - Nianci Zhu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing, 210009, China; Chongqing Innovation Institute of China Pharmaceutical University, Chongqing, 401135, China
| | - Sijia Chen
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing, 210009, China; Chongqing Innovation Institute of China Pharmaceutical University, Chongqing, 401135, China
| | - Meng Jia
- Nanjing University, School of Life Sciences, Nanjing, 210093, China
| | - Lingjing Xue
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing, 210009, China; Chongqing Innovation Institute of China Pharmaceutical University, Chongqing, 401135, China
| | - Meixi Hao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing, 210009, China; Chongqing Innovation Institute of China Pharmaceutical University, Chongqing, 401135, China.
| | - Can Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing, 210009, China; Chongqing Innovation Institute of China Pharmaceutical University, Chongqing, 401135, China.
| |
Collapse
|
87
|
Kim HR, Park JS, Soh WC, Kim NY, Moon HY, Lee JS, Jun CD. T Cell Microvilli: Finger-Shaped External Structures Linked to the Fate of T Cells. Immune Netw 2023; 23:e3. [PMID: 36911802 PMCID: PMC9995986 DOI: 10.4110/in.2023.23.e3] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 02/11/2023] [Accepted: 02/11/2023] [Indexed: 03/07/2023] Open
Abstract
Microvilli are outer membrane organelles that contain cross-linked filamentous actin. Unlike well-characterized epithelial microvilli, T-cell microvilli are dynamic similar to those of filopodia, which grow and shrink intermittently via the alternate actin-assembly and -disassembly. T-cell microvilli are specialized for sensing Ags on the surface of Ag-presenting cells (APCs). Thus, these finger-shaped microprotrusions contain many signaling-related proteins and can serve as a signaling platforms that induce intracellular signals. However, they are not limited to sensing external information but can provide sites for parts of the cell-body to tear away from the cell. Cells are known to produce many types of extracellular vesicles (EVs), such as exosomes, microvesicles, and membrane particles. T cells also produce EVs, but little is known about under what conditions T cells generate EVs and which types of EVs are released. We discovered that T cells produce few exosomes but release large amounsts of microvilli-derived particles during physical interaction with APCs. Although much is unanswered as to why T cells use the same organelles to sense Ags or to produce EVs, these events can significantly affect T cell fate, including clonal expansion and death. Since TCRs are localized at microvilli tips, this membrane event also raises a new question regarding long-standing paradigm in T cell biology; i.e., surface TCR downmodulation following T cell activation. Since T-cell microvilli particles carry T-cell message to their cognate partner, these particles are termed T-cell immunological synaptosomes (TISs). We discuss the potential physiological role of TISs and their application to immunotherapies.
Collapse
Affiliation(s)
- Hye-Ran Kim
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
- Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
- Division of Rare and Refractory Cancer, Tumor Immunology, Research Institute, National Cancer Center, Goyang 10408, Korea
| | - Jeong-Su Park
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
- Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
| | - Won-Chang Soh
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
- Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
| | - Na-Young Kim
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
- Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
| | - Hyun-Yoong Moon
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
- Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
| | - Ji-Su Lee
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
- Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
| | - Chang-Duk Jun
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
- Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
| |
Collapse
|
88
|
Espie D, Donnadieu E. CAR T-cell behavior and function revealed by real-time imaging. Semin Immunopathol 2023; 45:229-239. [PMID: 36688965 DOI: 10.1007/s00281-023-00983-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 01/03/2023] [Indexed: 01/24/2023]
Abstract
Adoptive transfer of T-cells expressing chimeric antigen receptors (CAR) has shown remarkable clinical efficacy against advanced B-cell malignancies. Nonetheless, the field of CAR T-cells is currently facing several major challenges. In particular, the CAR T-cell strategy has not yet produced favorable clinical responses when targeting solid tumors. In this context, it is of paramount importance to understand the determinants that limit the efficacy of T-cell-based immunotherapy. Characterization of CAR T-cells is usually based on flow cytometry and whole-transcriptome profiling. These approaches have been very valuable to determine intrinsic elements that condition T-cell ability to proliferate and expand. However, they do not take into account spatial and kinetic aspects of T-cell responses. In particular, in order to control tumor growth, CAR T-cells need to enter into the tumor, migrate within a complex tumor environment, and form productive conjugates with their targets. Advanced imaging techniques combined with innovative preclinical models represent promising tools to uncover the dynamics of CAR T-cells. In this review, we will discuss recent results on the biology of engineered T-cells that have been obtained with real-time imaging microscopy. Important notions have emerged from these imaging-based studies, such as the multi-killing potential of CAR T-cells. Finally, we will highlight how imaging techniques combined with other tools can solve remaining unresolved questions in the field of engineered T-cells.
Collapse
Affiliation(s)
- David Espie
- Université Paris Cité, CNRS, INSERM, Equipe Labellisée Ligue Contre le Cancer, Institut Cochin, INSERM U1016, 22 rue Méchain, F-75014, Paris, France.,Invectys, Paris, France
| | - Emmanuel Donnadieu
- Université Paris Cité, CNRS, INSERM, Equipe Labellisée Ligue Contre le Cancer, Institut Cochin, INSERM U1016, 22 rue Méchain, F-75014, Paris, France.
| |
Collapse
|
89
|
Cerny O. Quantification of interaction frequency between antigen-presenting cells and T cells by conjugation assay. Methods Cell Biol 2023; 173:65-75. [PMID: 36653086 DOI: 10.1016/bs.mcb.2022.07.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Interaction between an antigen-presenting cell and a T cell, and their subsequent conjugation are a prerequisite for the formation of the immunological synapse and productive, antigen-dependent activation of T cells. This initial interaction is accompanied by recognition of the presented antigen by the T cell receptor, and by changes in the morphology of the interacting cells and in actin cytoskeleton structure in the site of interaction. The experimental protocol below describes a simple assay for quantitative assessment of antigen-presenting cells-T cell conjugation using confocal microscopy or flow cytometry.
Collapse
Affiliation(s)
- Ondrej Cerny
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic.
| |
Collapse
|
90
|
Kim HR, Park JS, Kim NY, Jun CD. T Cell Immunological Synaptosomes: Definition and Isolation. Methods Mol Biol 2023; 2654:201-215. [PMID: 37106184 DOI: 10.1007/978-1-0716-3135-5_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
In addition to microvilli's role as structural scaffold for TCR clustering, we recently discovered a novel function as message senders. We found that microvilli are separated from the T cell body shortly upon TCR stimulation and vesiculated to form T cell microvilli particles (TMPs), a new type of membrane vesicles. TMPs and synaptic ectosomes, which bud from the synaptic cleft, constitute "T cell immunological synaptosomes (TISs)" and act as conveyors of T cell messages or traits to cognate antigen-presenting cells. In practice, it is almost impossible to distinguish between TMPs and synaptic ectosomes. Here, we describe a newly developed protocol to isolate TISs from activated T cells using antibody-immobilized agarose beads and density gradient ultracentrifugation. We further describe the methods for TIS quantification with flow cytometry and to evaluate TIS efficacy on dendritic cells.
Collapse
Affiliation(s)
- Hye-Ran Kim
- School of Life Sciences, Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, South Korea
- Division of Rare and Refractory Cancer, Immuno-oncology, Research Institute, National Cancer Center, Goyang, South Korea
| | - Jeong-Su Park
- School of Life Sciences, Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, South Korea
| | - Na-Young Kim
- School of Life Sciences, Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, South Korea
| | - Chang-Duk Jun
- School of Life Sciences, Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, South Korea.
| |
Collapse
|
91
|
Gan J, Cho JH, Lee R, Naghizadeh A, Poon LY, Wang E, Hui Z, Liu D. Methods of Machine Learning-Based Chimeric Antigen Receptor Immunological Synapse Quality Quantification. Methods Mol Biol 2023; 2654:493-502. [PMID: 37106203 PMCID: PMC10812845 DOI: 10.1007/978-1-0716-3135-5_32] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
Chimeric Antigen Receptor (CAR)-mediated immunotherapy shows promising results for refractory blood cancers. Currently, six CAR-T drugs have been approved by U.S. Food and Drug Administration (FDA). Theoretically, CAR-T cells must form an effective immunological synapse (IS, an interface between effective cells and their target cells) with their susceptible tumor cells to eliminate tumor cells. Previous studies show that CAR IS quality can be used as a predictive functional biomarker for CAR-T immunotherapies. However, quantification of CAR-T IS quality is clinically challenging. Machine learning (ML)-based CAR-T IS quality quantification has been proposed previously.Here, we show an easy-to-use, step-by-step approach to predicting the efficacy of CAR-modified cells using ML-based CAR IS quality quantification. This approach will guide the users on how to use ML-based CAR IS quality quantification in detail, which include: how to image CAR IS on the glass-supported planar lipid bilayer, how to define the CAR IS focal plane, how to segment the CAR IS images, and how to quantify the IS quality using ML-based algorithms.This approach will significantly enhance the accuracy and proficiency of CAR IS prediction in research.
Collapse
Affiliation(s)
- Julian Gan
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers University-New Jersey Medical School, Newark, NJ, USA
| | - Jong Hyun Cho
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers University-New Jersey Medical School, Newark, NJ, USA
- Center for Immunity and Inflammation, New Jersey Medical School, Newark, NJ, USA
| | - Ryan Lee
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers University-New Jersey Medical School, Newark, NJ, USA
| | - Alireza Naghizadeh
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers University-New Jersey Medical School, Newark, NJ, USA
| | - Ling Yue Poon
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers University-New Jersey Medical School, Newark, NJ, USA
| | - Ethan Wang
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers University-New Jersey Medical School, Newark, NJ, USA
| | - Zachary Hui
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers University-New Jersey Medical School, Newark, NJ, USA
| | - Dongfang Liu
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers University-New Jersey Medical School, Newark, NJ, USA.
- Center for Immunity and Inflammation, New Jersey Medical School, Newark, NJ, USA.
| |
Collapse
|
92
|
Abdelrahman A, Smith AS, Sengupta K. Observing Membrane and Cell Adhesion via Reflection Interference Contrast Microscopy. Methods Mol Biol 2023; 2654:123-135. [PMID: 37106179 DOI: 10.1007/978-1-0716-3135-5_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
Reflection interference contrast microscopy (RICM) is an optical microscopy technique ideally suited for imaging adhesion. While RICM (and the closely related interference reflection microscopy (IRM)) has been extensively used qualitatively or semiquantitatively to image cells, including immune cells, it can also be used quantitatively to measure membrane to surface distance, especially for model membranes. Here, we present a protocol for RICM and IRM imaging and the details of semiquantitative and quantitative analysis.
Collapse
Affiliation(s)
- Ahmed Abdelrahman
- Aix Marseille University, CNRS, CINAM, Turing Centre for Living Systems, Marseille, France
| | - Ana-Sunčana Smith
- PULS Group, Department of Physics, Centre for Computational Materials and Processes, Friedrich Alexander University Erlangen-Nürnberg, IZNF, Erlangen, Germany.
- Group for Computational Life Sciences, Division of Physical Chemistry, Ruđer Bošković Institute, Zagreb, Croatia.
| | - Kheya Sengupta
- Aix Marseille University, CNRS, CINAM, Turing Centre for Living Systems, Marseille, France
| |
Collapse
|
93
|
Mørch AM, Schneider F. Investigating Diffusion Dynamics and Interactions with Scanning Fluorescence Correlation Spectroscopy (sFCS). Methods Mol Biol 2023; 2654:61-89. [PMID: 37106176 DOI: 10.1007/978-1-0716-3135-5_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
Activation of immune cells and formation of immunological synapses (IS) rely critically on the reorganization of the plasma membrane. These highly orchestrated processes are driven by diffusion and oligomerization dynamics, as well as by single molecule interactions. While slow macro- and meso-scale changes in organization can be observed with conventional imaging, fast nano-scale dynamics are often missed with traditional approaches, but resolving them is, nonetheless, essential to understand the underlying biological mechanisms at play. Here, we describe the use of scanning fluorescence correlation spectroscopy (sFCS) and scanning fluorescence cross-correlation spectroscopy (sFCCS) to study reorganization and changes in molecular diffusion dynamics and interactions during IS formation and in other biological settings. We focus on the practical aspects of the measurements including calibration and alignment of the optical setup, present a comprehensive protocol to perform the measurements, and provide data analysis pipelines and strategies. Finally, we show an exemplary application of the technology to studying Lck diffusion during T-cell signaling.
Collapse
Affiliation(s)
| | - Falk Schneider
- Translational Imaging Center, University of Southern California, Los Angeles, California, USA.
| |
Collapse
|
94
|
Lanna A, D'Ambra C. Detection of Telomere Transfer at Immunological Synapse. Methods Mol Biol 2023; 2654:251-261. [PMID: 37106187 DOI: 10.1007/978-1-0716-3135-5_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
Eukaryotes solve the DNA-end replication problem synthesizing hexameric chromosome ends known as telomeres. Recent studies have uncovered unexpected functions of telomeres in linking synaptic signaling and vesicle transport, with at least one pathway directly involved in transferring telomeres through the immune synapse. These emerging forms of cellular communication may originate a new class of antiaging interventions based on telomere transplants.
Collapse
|
95
|
Alatoom A, ElGindi M, Sapudom J, Teo JCM. The T Cell Journey: A Tour de Force. Adv Biol (Weinh) 2023; 7:e2200173. [PMID: 36190140 DOI: 10.1002/adbi.202200173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/30/2022] [Indexed: 11/07/2022]
Abstract
T cells act as the puppeteers in the adaptive immune response, and their dysfunction leads to the initiation and progression of pathological conditions. During their lifetime, T cells experience myriad forces that modulate their effector functions. These forces are imposed by interacting cells, surrounding tissues, and shear forces from fluid movement. In this review, a journey with T cells is made, from their development to their unique characteristics, including the early studies that uncovered their mechanosensitivity. Then the studies pertaining to the responses of T cell activation to changes in antigen-presenting cells' physical properties, to their immediate surrounding extracellular matrix microenvironment, and flow conditions are highlighted. In addition, it is explored how pathological conditions like the tumor microenvironment can hinder T cells and allow cancer cells to escape elimination.
Collapse
Affiliation(s)
- Aseel Alatoom
- Laboratory for Immuno Bioengineering Research and Applications Division of Engineering, New York University Abu Dhabi, Saadiyat Campus, P.O. Box 127788, Abu Dhabi, UAE.,Department of Mechanical Engineering Tandon School of Engineering, New York University, 6 MetroTech Center, Brooklyn, NY, 11201, USA
| | - Mei ElGindi
- Laboratory for Immuno Bioengineering Research and Applications Division of Engineering, New York University Abu Dhabi, Saadiyat Campus, P.O. Box 127788, Abu Dhabi, UAE
| | - Jiranuwat Sapudom
- Laboratory for Immuno Bioengineering Research and Applications Division of Engineering, New York University Abu Dhabi, Saadiyat Campus, P.O. Box 127788, Abu Dhabi, UAE
| | - Jeremy C M Teo
- Laboratory for Immuno Bioengineering Research and Applications Division of Engineering, New York University Abu Dhabi, Saadiyat Campus, P.O. Box 127788, Abu Dhabi, UAE.,Department of Mechanical Engineering Tandon School of Engineering, New York University, 6 MetroTech Center, Brooklyn, NY, 11201, USA.,Department of Biomedical Engineering Tandon School of Engineering, New York University, 6 MetroTech Center, Brooklyn, NY, 11201, USA
| |
Collapse
|
96
|
Abstract
Here, we describe a method, which we term "shadow imaging," to analyze the secretions of individual cells at immune synapses or other cell contacts. Following immune synapse formation and cellular activation on ligand-rich slides, the position of each cell is recorded using a pulsed immunofluorescence stain against the proteins on the ligand-rich slide surface. The pulsed stain does not penetrate the synaptic cleft, resulting in an unlabeled region or "shadow" beneath cells that is retained following cellular detachment. The secreted components, such as perforin, exosomes, or other types of extracellular vesicles, are retained on the slide and can be analyzed on a single-cell basis using immunofluorescence. The ability to identify single cells secreting different combinations of particles, proteins, and vesicles enables us to better understand the heterogeneity in immune cell secretions and can be used as a novel approach for phenotyping cell populations.
Collapse
Affiliation(s)
- Ashley R Ambrose
- The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - Khodor S Hazime
- The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
- Department of Life Sciences, Imperial College London, London, UK
| | - Daniel M Davis
- The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK.
- Department of Life Sciences, Imperial College London, London, UK.
| |
Collapse
|
97
|
Bispecific Antibody Format and the Organization of Immunological Synapses in T Cell-Redirecting Strategies for Cancer Immunotherapy. Pharmaceutics 2022; 15:pharmaceutics15010132. [PMID: 36678761 PMCID: PMC9863865 DOI: 10.3390/pharmaceutics15010132] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/16/2022] [Accepted: 12/24/2022] [Indexed: 01/03/2023] Open
Abstract
T cell-redirecting strategies have emerged as effective cancer immunotherapy approaches. Bispecific antibodies (bsAbs) are designed to specifically recruit T cells to the tumor microenvironment and induce the assembly of the immunological synapse (IS) between T cells and cancer cells or antigen-presenting cells. The way that the quality of the IS might predict the effectiveness of T cell-redirecting strategies, including those mediated by bsAbs or by chimeric antigen receptors (CAR)-T cells, is currently under discussion. Here we review the organization of the canonical IS assembled during natural antigenic stimulation through the T cell receptor (TCR) and to what extent different bsAbs induce T cell activation, canonical IS organization, and effector function. Then, we discuss how the biochemical parameters of different formats of bsAbs affect the effectivity of generating an antigen-induced canonical IS. Finally, the quality of the IS assembled by bsAbs and monoclonal antibodies or CAR-T cells are compared, and strategies to improve bsAb-mediated T cell-redirecting strategies are discussed.
Collapse
|
98
|
McAffee DB, O'Dair MK, Lin JJ, Low-Nam ST, Wilhelm KB, Kim S, Morita S, Groves JT. Discrete LAT condensates encode antigen information from single pMHC:TCR binding events. Nat Commun 2022; 13:7446. [PMID: 36460640 PMCID: PMC9718779 DOI: 10.1038/s41467-022-35093-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 11/17/2022] [Indexed: 12/05/2022] Open
Abstract
LAT assembly into a two-dimensional protein condensate is a prominent feature of antigen discrimination by T cells. Here, we use single-molecule imaging techniques to resolve the spatial position and temporal duration of each pMHC:TCR molecular binding event while simultaneously monitoring LAT condensation at the membrane. An individual binding event is sufficient to trigger a LAT condensate, which is self-limiting, and neither its size nor lifetime is correlated with the duration of the originating pMHC:TCR binding event. Only the probability of the LAT condensate forming is related to the pMHC:TCR binding dwell time. LAT condenses abruptly, but after an extended delay from the originating binding event. A LAT mutation that facilitates phosphorylation at the PLC-γ1 recruitment site shortens the delay time to LAT condensation and alters T cell antigen specificity. These results identify a function for the LAT protein condensation phase transition in setting antigen discrimination thresholds in T cells.
Collapse
Affiliation(s)
- Darren B McAffee
- Department of Chemistry, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Mark K O'Dair
- Department of Chemistry, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Jenny J Lin
- Department of Chemistry, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Shalini T Low-Nam
- Department of Chemistry, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Kiera B Wilhelm
- Department of Chemistry, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Sungi Kim
- Department of Chemistry, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Shumpei Morita
- Department of Chemistry, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Jay T Groves
- Department of Chemistry, University of California, Berkeley, Berkeley, CA, 94720, USA.
- Institute for Digital Molecular Analytics and Science, Nanyang Technological University, 59 Nanyang Drive, Singapore, 636921, Singapore.
| |
Collapse
|
99
|
Teppert K, Wang X, Anders K, Evaristo C, Lock D, Künkele A. Joining Forces for Cancer Treatment: From "TCR versus CAR" to "TCR and CAR". Int J Mol Sci 2022; 23:14563. [PMID: 36498890 PMCID: PMC9739809 DOI: 10.3390/ijms232314563] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/14/2022] [Accepted: 11/19/2022] [Indexed: 11/24/2022] Open
Abstract
T cell-based immunotherapy has demonstrated great therapeutic potential in recent decades, on the one hand, by using tumor-infiltrating lymphocytes (TILs) and, on the other hand, by engineering T cells to obtain anti-tumor specificities through the introduction of either engineered T cell receptors (TCRs) or chimeric antigen receptors (CARs). Given the distinct design of both receptors and the type of antigen that is encountered, the requirements for proper antigen engagement and downstream signal transduction by TCRs and CARs differ. Synapse formation and signal transduction of CAR T cells, despite further refinement of CAR T cell designs, still do not fully recapitulate that of TCR T cells and might limit CAR T cell persistence and functionality. Thus, deep knowledge about the molecular differences in CAR and TCR T cell signaling would greatly advance the further optimization of CAR designs and elucidate under which circumstances a combination of both receptors would improve the functionality of T cells for cancer treatment. Herein, we provide a comprehensive review about similarities and differences by directly comparing the architecture, synapse formation and signaling of TCRs and CARs, highlighting the knowns and unknowns. In the second part of the review, we discuss the current status of combining CAR and TCR technologies, encouraging a change in perspective from "TCR versus CAR" to "TCR and CAR".
Collapse
Affiliation(s)
- Karin Teppert
- Miltenyi Biotec B.V. & Co. KG, 51429 Bergisch Gladbach, Germany
| | - Xueting Wang
- Miltenyi Biotec B.V. & Co. KG, 51429 Bergisch Gladbach, Germany
| | - Kathleen Anders
- German Cancer Consortium (DKTK), 10117 Berlin, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - César Evaristo
- Miltenyi Biotec B.V. & Co. KG, 51429 Bergisch Gladbach, Germany
| | - Dominik Lock
- Miltenyi Biotec B.V. & Co. KG, 51429 Bergisch Gladbach, Germany
| | - Annette Künkele
- German Cancer Consortium (DKTK), 10117 Berlin, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Department of Pediatric Oncology and Hematology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany
| |
Collapse
|
100
|
Wen Y, Ma J. Phase separation drives the formation of biomolecular condensates in the immune system. Front Immunol 2022; 13:986589. [PMID: 36439121 PMCID: PMC9685520 DOI: 10.3389/fimmu.2022.986589] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 10/19/2022] [Indexed: 08/12/2023] Open
Abstract
When the external conditions change, such as the temperature or the pressure, the multi-component system sometimes separates into several phases with different components and structures, which is called phase separation. Increasing studies have shown that cells condense related biomolecules into independent compartments in order to carry out orderly and efficient biological reactions with the help of phase separation. Biomolecular condensates formed by phase separation play a significant role in a variety of cellular processes, including the control of signal transduction, the regulation of gene expression, and the stress response. In recent years, many phase separation events have been discovered in the immune response process. In this review, we provided a comprehensive and detailed overview of the role and mechanism of phase separation in the innate and adaptive immune responses, which will help the readers to appreciate the advance and importance of this field.
Collapse
Affiliation(s)
- Yuqing Wen
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Cancer Research Institute and School of Basic Medical Science, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Hunan Key Laboratory of Cancer Metabolism, Changsha, China
| | - Jian Ma
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Cancer Research Institute and School of Basic Medical Science, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Hunan Key Laboratory of Cancer Metabolism, Changsha, China
| |
Collapse
|